176 results on '"Leon J. De Windt"'
Search Results
2. Quantify permeability using on-a-chip models in high-throughput applications
- Author
-
Camilla Soragni, Tessa Vergroesen, Nynke Hettema, Gwenaëlle Rabussier, Henriëtte L. Lanz, Sebastian J. Trietsch, Leon J. de Windt, and Chee P. Ng
- Subjects
Cell-based Assays ,High-throughput Screening ,Microscopy ,Biotechnology and bioengineering ,Science (General) ,Q1-390 - Abstract
Summary: Traditionally, to quantify permeability of a biological barrier, the initial slope is used, based on the assumption of sink condition (concentration of the donor is constant, and the receiver increases less than 10%). With on-a-chip barrier models, this assumption fails in cell-free or leaky conditions, which requires the use of the exact solution. To encounter a time delay from performing the assay and acquiring the data, we present a protocol with the exact equation modified to incorporate a time offset. : Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
- Published
- 2023
- Full Text
- View/download PDF
3. A versatile multiplexed assay to quantify intracellular ROS and cell viability in 3D on-a-chip models
- Author
-
Camilla Soragni, Gwenaëlle Rabussier, Henriëtte L. Lanz, Kristin M. Bircsak, Leon J. de Windt, Sebastiaan J. Trietsch, Colin E. Murdoch, and Chee Ping Ng
- Subjects
ROS ,Cell viability ,Multiplex assay ,Organ-on-a-chip ,Antioxidant ,Oxidative stress ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Reactive oxygen species (ROS) have different properties and biological functions. They contribute to cell signaling and, in excessive amounts, to oxidative stress (OS). Although ROS is pivotal in a wide number of physiological systems and pathophysiological processes, direct quantification in vivo is quite challenging and mainly limited to in vitro studies. Even though advanced in vitro cell culture techniques, like on-a-chip culture, have overcome the lack of crucial in vivo-like physiological aspects in 2D culture, the majority of in vitro ROS quantification studies are generally performed in 2D. Here we report the development, application, and validation of a multiplexed assay to quantify ROS and cell viability in organ-on-a-chip models. The assay utilizes three dyes to stain live cells for ROS, dead cells, and DNA. Confocal images were analyzed to quantify ROS probes and determine the number of nuclei and dead cells. We found that, in contrast to what has been reported with 2D cell culture, on-a-chip models are more prone to scavenge ROS rather than accumulate them. The assay is sensitive enough to distinguish between different phenotypes of endothelial cells (ECs) based on the level of OS to detect higher level in tumor than normal cells. Our results indicate that the use of physiologically relevant models and this assay could help unravelling the mechanisms behind OS and ROS accumulation. A further step could be taken in data analysis by implementing AI in the pipeline to also analyze images for morphological changes to have an even broader view of OS mechanism.
- Published
- 2022
- Full Text
- View/download PDF
4. A microRNA program regulates the balance between cardiomyocyte hyperplasia and hypertrophy and stimulates cardiac regeneration
- Author
-
Andrea Raso, Ellen Dirkx, Vasco Sampaio-Pinto, Hamid el Azzouzi, Ryan J. Cubero, Daniel W. Sorensen, Lara Ottaviani, Servé Olieslagers, Manon M. Huibers, Roel de Weger, Sailay Siddiqi, Silvia Moimas, Consuelo Torrini, Lorena Zentillin, Luca Braga, Diana S. Nascimento, Paula A. da Costa Martins, Jop H. van Berlo, Serena Zacchigna, Mauro Giacca, and Leon J. De Windt
- Subjects
Science - Abstract
Myocardial regeneration and proliferation of heart muscle cells is limited to a short period after birth early postnatal life, after which heart muscle cells can only grow in size and not in number. Here, the authors identified that the expression level of an endogenous microRNA cluster in heart muscle promotes the passage of the proliferative state to adult heart growth, and modulating the expression of this cluster can stimulate heart regeneration after myocardial infarction.
- Published
- 2021
- Full Text
- View/download PDF
5. Circulating miR-216a as a biomarker of metabolic alterations and obesity in women
- Author
-
Indira G.C. Vonhögen, Zenab Mohseni, Bjorn Winkens, Ke Xiao, Thomas Thum, Martina Calore, Paula A. da Costa Martins, Leon J. de Windt, Marc E.A. Spaanderman, and Chahinda Ghossein-Doha
- Subjects
microRNA ,Obesity ,Metabolic syndrome ,Biomarker ,Genetics ,QH426-470 - Abstract
Obesity leads to an amplified risk of disease and contributes to the occurrence of type 2 diabetes, fatty liver disease, coronary heart disease, stroke, chronic kidney disease and various types of cancer. MicroRNAs (miRNAs), small non-coding RNA molecules of 20–25 nucleotides, can remain stable in plasma and have been studied as potential (predictive) biomarkers for obesity and related metabolic disorders. The aim of this study was to identify circulating miRNAs as biomarkers for obesity status and metabolic alterations in women. Circulating miR-216a and miR-155–5p were selected by miRNA expression profiling and validated by real time quantitative PCR in a validation cohort of 60 obese women and 60 normal weight-age-matched control women. This was supplemented by correlation analysis of the candidate miRNA and anthropometric variables, blood biochemistry and lipid profile markers. Circulating miR-216a was validated as a biomarker of obesity status with significantly reduced levels in obese women. Interestingly, this was associated with a negative correlation between the plasma miR-216a content and body mass index (BMI), waist circumference, mean arterial pressure (MAP), triglycerides, ratio of total cholesterol/high density lipoprotein (HDL)-cholesterol and high sensitivity-C reactive protein (hs-CRP).Taken together, we provide evidence for an abnormally expressed circulating miRNA, miR-216a, with additive value as a predictive marker for obesity that correlates with metabolic alterations presented by lipid profile and inflammatory markers.
- Published
- 2020
- Full Text
- View/download PDF
6. Author Correction: A microRNA program regulates the balance between cardiomyocyte hyperplasia and hypertrophy and stimulates cardiac regeneration
- Author
-
Andrea Raso, Ellen Dirkx, Vasco Sampaio-Pinto, Hamid el Azzouzi, Ryan J. Cubero, Daniel W. Sorensen, Lara Ottaviani, Servé Olieslagers, Manon M. Huibers, Roel de Weger, Sailay Siddiqi, Silvia Moimas, Consuelo Torrini, Lorena Zentillin, Luca Braga, Diana S. Nascimento, Paula A. da Costa Martins, Jop H. van Berlo, Serena Zacchigna, Mauro Giacca, and Leon J. De Windt
- Subjects
Science - Published
- 2022
- Full Text
- View/download PDF
7. Extracellular Vesicle miRNAs in the Promotion of Cardiac Neovascularisation
- Author
-
Despoina Kesidou, Paula A. da Costa Martins, Leon J. de Windt, Mairi Brittan, Abdelaziz Beqqali, and Andrew Howard Baker
- Subjects
extracellular vesicles (EV) ,microRNA (miR) ,neovascularisation ,angiogenesis ,cardiac ,myocardial infarct ,Physiology ,QP1-981 - Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide claiming almost 17. 9 million deaths annually. A primary cause is atherosclerosis within the coronary arteries, which restricts blood flow to the heart muscle resulting in myocardial infarction (MI) and cardiac cell death. Despite substantial progress in the management of coronary heart disease (CHD), there is still a significant number of patients developing chronic heart failure post-MI. Recent research has been focused on promoting neovascularisation post-MI with the ultimate goal being to reduce the extent of injury and improve function in the failing myocardium. Cardiac cell transplantation studies in pre-clinical models have shown improvement in cardiac function; nonetheless, poor retention of the cells has indicated a paracrine mechanism for the observed improvement. Cell communication in a paracrine manner is controlled by various mechanisms, including extracellular vesicles (EVs). EVs have emerged as novel regulators of intercellular communication, by transferring molecules able to influence molecular pathways in the recipient cell. Several studies have demonstrated the ability of EVs to stimulate angiogenesis by transferring microRNA (miRNA, miR) molecules to endothelial cells (ECs). In this review, we describe the process of neovascularisation and current developments in modulating neovascularisation in the heart using miRNAs and EV-bound miRNAs. Furthermore, we critically evaluate methods used in cell culture, EV isolation and administration.
- Published
- 2020
- Full Text
- View/download PDF
8. Circulating miR-185-5p as a Potential Biomarker for Arrhythmogenic Right Ventricular Cardiomyopathy
- Author
-
Claudia Sacchetto, Zenab Mohseni, Robin M. W. Colpaert, Libero Vitiello, Marzia De Bortoli, Indira G. C. Vonhögen, Ke Xiao, Giulia Poloni, Alessandra Lorenzon, Chiara Romualdi, Riccardo Bariani, Elisa Mazzotti, Luciano Daliento, Barbara Bauce, Domenico Corrado, Thomas Thum, Alessandra Rampazzo, Leon J. de Windt, and Martina Calore
- Subjects
arrhythmogenic right ventricular cardiomyopathy ,MicroRNAs ,circulating microRNAs ,heart failure ,biomarkers ,genetics ,Cytology ,QH573-671 - Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a genetic cardiac disease characterized by progressive myocardial fibro-fatty replacement, arrhythmias and risk of sudden death. Its diagnosis is challenging and often it is achieved after disease onset or postmortem. In this study, we sought to identify circulating microRNAs (miRNAs) differentially expressed in ARVC patients compared to healthy controls. In the pilot study, we screened the expression of 754 miRNAs from 21 ARVC patients and 20 healthy controls. After filtering the miRNAs considering a log fold-change cut-off of ±1, p-value < 0.05, we selected five candidate miRNAs for a subsequent validation study in which we used TaqMan-based real-time PCR to analyse samples from 37 ARVC patients and 30 healthy controls. We found miR-185-5p significantly upregulated in ARVC patients. Receiver operating characteristic analysis indicated an area under the curve of 0.854, corroborating the link of this miRNA and ARVC pathophysiology.
- Published
- 2021
- Full Text
- View/download PDF
9. An unbiased silencing screen in muscle cells identifies miR-320a, miR-150, miR-196b, and miR-34c as regulators of skeletal muscle mitochondrial metabolism
- Author
-
Dennis Dahlmans, Alexandre Houzelle, Pénélope Andreux, Johanna A. Jörgensen, Xu Wang, Leon J. de Windt, Patrick Schrauwen, Johan Auwerx, and Joris Hoeks
- Subjects
Skeletal muscle metabolism ,Mitochondria ,MicroRNA ,Screening ,Type 2 diabetes ,Oxidative capacity ,Internal medicine ,RC31-1245 - Abstract
Objective: Strategies improving skeletal muscle mitochondrial capacity are commonly paralleled by improvements in (metabolic) health. We and others previously identified microRNAs regulating mitochondrial oxidative capacity, but data in skeletal muscle are limited. Therefore, the present study aimed to identify novel microRNAs regulating skeletal muscle mitochondrial metabolism. Methods and results: We conducted an unbiased, hypothesis-free microRNA silencing screen in C2C12 myoblasts, using >700 specific microRNA inhibitors, and investigated a broad panel of mitochondrial markers. After subsequent validation in differentiated C2C12 myotubes, and exclusion of microRNAs without a human homologue or with an adverse effect on mitochondrial metabolism, 19 candidate microRNAs remained. Human clinical relevance of these microRNAs was investigated by measuring their expression in human skeletal muscle of subject groups displaying large variation in skeletal muscle mitochondrial capacity. Conclusion: The results show that that microRNA-320a, microRNA-196b-3p, microRNA-150-5p, and microRNA-34c-3p are tightly related to in vivo skeletal muscle mitochondrial function in humans and identify these microRNAs as targets for improving mitochondrial metabolism.
- Published
- 2017
- Full Text
- View/download PDF
10. miR-199b-5p is a regulator of left ventricular remodeling following myocardial infarction
- Author
-
Burcu Duygu, Ella M. Poels, Rio Juni, Nicole Bitsch, Lara Ottaviani, Servé Olieslagers, Leon J. de Windt, and Paula A. da Costa Martins
- Subjects
Genetics ,QH426-470 - Abstract
Myocardial infarction (MI), the globally leading cause of heart failure, morbidity and mortality, involves post-MI ventricular remodeling, a complex process including acute injury healing, scar formation and global changes in the surviving myocardium. The molecular mechanisms involved in adverse post-infarct left ventricular remodeling still remain poorly defined. Recently, microRNAs have been implicated in the development and progression of various cardiac diseases as crucial regulators of gene expression. We previously demonstrated that in a murine model of pressure overload, a model of heart failure secondary to aortic stenosis or chronic high blood pressure, elevated myocardial expression of miR-199b-5p is sufficient to activate calcineurin/NFAT signaling, leading to exaggerated cardiac pathological remodeling and dysfunction. Given the differences in left ventricular remodeling secondary to post-infarct healing and pressure overload, we evaluated miR-199b function in post-MI remodeling. We confirmed that the expression of miR-199b is elevated in the post-infarcted heart. Transgenic animals with cardiomyocyte-restricted overexpression of miR-199b-5p displayed exaggerated pathological remodeling after MI, reflected by severe systolic and diastolic dysfunction and fibrosis deposition. Conversely, therapeutic silencing of miR-199b-5p in MI-induced cardiac remodeling by using an antagomir to specifically inhibit endogenous miR-199b-5p in vivo, resulted in efficient suppression of cardiac miR-199b-5p expression and attenuated cardiac dysfunction and dilation following MI. Mechanistically, miR-199b-5p influenced the expression of three predicted target genes in post-infarcted hearts, dual specificity tyrosine-phosphorylation-regulated kinase 1A (Dyrk1a), the notch1 receptor and its ligand jagged1. In conclusion, here we provide evidence supporting that stress-induced miR-199b-5p participates in post-infarct remodeling by simultaneous regulation of distinct target genes.
- Published
- 2017
- Full Text
- View/download PDF
11. Non-coding RNA function in stem cells and Regenerative Medicine
- Author
-
Leon J. De Windt
- Subjects
Genetics ,QH426-470 - Published
- 2018
- Full Text
- View/download PDF
12. MiR-337-3p Promotes Adipocyte Browning by Inhibiting TWIST1
- Author
-
Indira G.C. Vonhögen, Hamid el Azzouzi, Servé Olieslagers, Aliaksei Vasilevich, Jan de Boer, Francisco J. Tinahones, Paula A. da Costa Martins, Leon J. de Windt, and Mora Murri
- Subjects
brown adipose tissue ,microRNA ,mitochondria ,obesity ,metabolic syndrome ,Cytology ,QH573-671 - Abstract
The prevalence of metabolic syndrome (MetS) and obesity is an alarming health issue worldwide. Obesity is characterized by an excessive accumulation of white adipose tissue (WAT), and it is associated with diminished brown adipose tissue (BAT) activity. Twist1 acts as a negative feedback regulator of BAT metabolism. Therefore, targeting Twist1 could become a strategy for obesity and metabolic disease. Here, we have identified miR-337-3p as an upstream regulator of Twist1. Increased miR-337-3p expression paralleled decreased expression of TWIST1 in BAT compared to WAT. Overexpression of miR-337-3p in brown pre-adipocytes provoked a reduction in Twist1 expression that was accompanied by increased expression of brown/mitochondrial markers. Luciferase assays confirmed an interaction between the miR-337 seed sequence and Twist1 3′UTR. The inverse relationship between the expression of TWIST1 and miR-337 was finally validated in adipose tissue samples from non-MetS and MetS subjects that demonstrated a dysregulation of the miR-337-Twist1 molecular axis in MetS. The present study demonstrates that adipocyte miR-337-3p suppresses Twist1 repression and enhances the browning of adipocytes.
- Published
- 2020
- Full Text
- View/download PDF
13. Cardiovascular extracellular microRNAs: emerging diagnostic markers and mechanisms of cell-to-cell RNA communication
- Author
-
Virginie Kinet, Julie Halkein, Ellen Dirkx, and Leon J. De Windt
- Subjects
extracellular microRNA ,inter-cellular communications ,biomarkers ,cardiovascular diseases ,Genetics ,QH426-470 - Abstract
Cardiovascular diseases are a leading cause of morbidity and mortality in Western societies. It is now well established that microRNAs (miRNAs) are determinant regulators in various medical conditions including cardiovascular diseases. The recent discovery that miRNAs, while associated with different carriers, can be exported out of the cell, has triggered a renewed interest to analyze the potential to use extracellular miRNAs as tools for diagnostic and therapeutic studies. Circulating miRNAs in biological fluids present a technological advantage compared to current diagnostic tools by virtue of their remarkable stability and relative ease of detection rendering them ideal tools for non-invasive and rapid diagnosis. Extracellular miRNAs also represent a novel form of inter-cellular communication by transferring genetic information from a donor cell to a recipient cell. This review briefly summarizes recent insights in the origin, function and diagnostic potential of extracellular miRNAs by focusing on a select number of cardiovascular diseases.
- Published
- 2013
- Full Text
- View/download PDF
14. Correction: A Deep Sequencing Approach to Uncover the miRNOME in the Human Heart.
- Author
-
Stefanos Leptidis, Hamid el Azzouzi, Sjoukje I. Lok, Roel de Weger, Servé Olieslagers, Natasja Kisters, Gustavo J. Silva, Stephane Heymans, Edwin Cuppen, Eugene Berezikov, Leon J. De Windt, and Paula da Costa Martins
- Subjects
Medicine ,Science - Published
- 2013
- Full Text
- View/download PDF
15. Healthy and diseased placental barrier on-a-chip models suitable for standardized studies
- Author
-
Gwenaëlle Rabussier, Ivan Bünter, Josse Bouwhuis, Camilla Soragni, Torben van Zijp, Chee Ping Ng, Karel Domansky, Leon J. de Windt, Paul Vulto, Colin E. Murdoch, Kristin M. Bircsak, and Henriëtte L. Lanz
- Subjects
Biomaterials ,Biomedical Engineering ,General Medicine ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2023
- Full Text
- View/download PDF
16. Strategies and challenges for non-viral delivery of non-coding RNAs to the heart
- Author
-
Andreia Vilaça, Leon J. de Windt, Hugo Fernandes, and Lino Ferreira
- Subjects
Molecular Medicine ,Molecular Biology - Abstract
Non-coding RNAs (ncRNAs), such as miRNAs and long non-coding RNAs (lncRNAs) have been reported as regulators of cardiovascular pathophysiology. Their transient effect and diversified mechanisms of action offer a plethora of therapeutic opportunities for cardiovascular diseases (CVDs). However, physicochemical RNA features such as charge, stability, and structural organization hinder efficient on-target cellular delivery. Here, we highlight recent preclinical advances in ncRNA delivery for the cardiovascular system using non-viral approaches. We identify the unmet needs and advance possible solutions towards clinical translation. Finding the optimal delivery vehicle and administration route is vital to improve therapeutic efficacy and safety; however, given the different types of ncRNAs, this may ultimately not be frameable within a one-size-fits-all approach.
- Published
- 2023
- Full Text
- View/download PDF
17. MicroRNA-216a is essential for cardiac angiogenesis
- Author
-
Rio P. Juni, Jordy M.M. Kocken, Ricardo C. Abreu, Lara Ottaviani, Tim Davalan, Burcu Duygu, Ella M. Poels, Aliaksei Vasilevich, Jana C. Hegenbarth, Mahesh Appari, Nicole Bitsch, Serve Olieslagers, Dorien M. Schrijvers, Monika Stoll, Joerg Heineke, Jan de Boer, Leon J. de Windt, Paula A. da Costa Martins, Physiology, ACS - Heart failure & arrhythmias, and ACS - Microcirculation
- Subjects
Pharmacology ,Drug Discovery ,Genetics ,Molecular Medicine ,Molecular Biology - Abstract
While it is experimentally supported that impaired myocardial vascularization contributes to a mismatch between myocardial oxygen demand and supply, a mechanistic basis for disruption of coordinated tissue growth and angiogenesis in heart failure remains poorly understood. Silencing strategies that impair microRNA biogenesis have firmly implicated microRNAs in the regulation of angiogenesis, and individual microRNAs prove to be crucial in developmental or tumor angiogenesis. A high-throughput functional screening for the analysis of a whole-genome microRNA silencing library with regard to their phenotypic effect on endothelial cell proliferation as a key parameter, revealed several anti- and pro-proliferative microRNAs. Among those was miR-216a, a pro-angiogenic microRNA which is enriched in cardiac microvascular endothelial cells and reduced in expression under cardiac stress conditions. miR-216a null mice display dramatic cardiac phenotypes related to impaired myocardial vascularization and unbalanced autophagy and inflammation, supporting a model where microRNA regulation of microvascularization impacts the cardiac response to stress.
- Published
- 2023
- Full Text
- View/download PDF
18. DNA repair in cardiomyocytes is critical for maintaining cardiac function in mice
- Author
-
Martine de Boer, Maaike te Lintel Hekkert, Jiang Chang, Bibi S. van Thiel, Leonie Martens, Maxime M. Bos, Marion G. J. de Kleijnen, Yanto Ridwan, Yanti Octavia, Elza D. van Deel, Lau A. Blonden, Renata M. C. Brandt, Sander Barnhoorn, Paula K. Bautista‐Niño, Ilona Krabbendam‐Peters, Rianne Wolswinkel, Banafsheh Arshi, Mohsen Ghanbari, Christian Kupatt, Leon J. de Windt, A. H. Jan Danser, Ingrid van der Pluijm, Carol Ann Remme, Monika Stoll, Joris Pothof, Anton J. M. Roks, Maryam Kavousi, Jeroen Essers, Jolanda van der Velden, Jan H. J. Hoeijmakers, Dirk J. Duncker, Cardiology, Experimental Cardiology, ACS - Heart failure & arrhythmias, APH - Methodology, RS: Carim - H05 Gene regulation, RS: FSE DMG, Biochemie, RS: Carim - B01 Blood proteins & engineering, Molecular Genetics, Epidemiology, Internal Medicine, Surgery, and Radiotherapy
- Subjects
DAMAGE ,Aging ,ROLES ,ABNORMALITIES ,PROTEINS ,apoptosis ,RECOMBINATION ,DNA repair ,Cell Biology ,BRCA1 ,CANCER ,congestive heart failure ,NUCLEOTIDE EXCISION-REPAIR ,HEART-FAILURE ,DNA damage ,cardiac function ,MYOCARDIUM - Abstract
Heart failure has reached epidemic proportions in a progressively ageing population. The molecular mechanisms underlying heart failure remain elusive, but evidence indicates that DNA damage is enhanced in failing hearts. Here, we tested the hypothesis that endogenous DNA repair in cardiomyocytes is critical for maintaining normal cardiac function, so that perturbed repair of spontaneous DNA damage drives early onset of heart failure. To increase the burden of spontaneous DNA damage, we knocked out the DNA repair endonucleases xeroderma pigmentosum complementation group G (XPG) and excision repair cross-complementation group 1 (ERCC1), either systemically or cardiomyocyte-restricted, and studied the effects on cardiac function and structure. Loss of DNA repair permitted normal heart development but subsequently caused progressive deterioration of cardiac function, resulting in overt congestive heart failure and premature death within 6 months. Cardiac biopsies revealed increased oxidative stress associated with increased fibrosis and apoptosis. Moreover, gene set enrichment analysis showed enrichment of pathways associated with impaired DNA repair and apoptosis, and identified TP53 as one of the top active upstream transcription regulators. In support of the observed cardiac phenotype in mutant mice, several genetic variants in the ERCC1 and XPG gene in human GWAS data were found to be associated with cardiac remodelling and dysfunction. In conclusion, unrepaired spontaneous DNA damage in differentiated cardiomyocytes drives early onset of cardiac failure. These observations implicate DNA damage as a potential novel therapeutic target and highlight systemic and cardiomyocyte-restricted DNA repair-deficient mouse mutants as bona fide models of heart failure.
- Published
- 2023
19. Evolutionarily conserved transcriptional landscape of the heart defining the chamber specific physiology
- Author
-
Federica De Majo, Ulrich Schotten, Leon J. De Windt, Monika Stoll, Shrey Gandhi, Anika Witten, Martijn Gilbers, Jos G. Maessen, RS: FSE DMG, RS: Carim - H08 Experimental atrial fibrillation, RS: Carim - V04 Surgical intervention, RS: Carim - B01 Blood proteins & engineering, Fysiologie, MUMC+: MA Med Staf Artsass CTC (9), CTC, MUMC+: MA Cardiothoracale Chirurgie (3), Biochemie, and RS: FHML MaCSBio
- Subjects
lncRNA conservation ,DATABASE ,ved/biology.organism_classification_rank.species ,Computational biology ,Biology ,Synteny ,LONG NONCODING RNAS ,Transcriptome ,Cardiac transcriptome ,Chamber specificity ,Transcription (biology) ,biology.animal ,ATRIAL ,Gene expression ,Genetics ,Model organism ,Gene ,GENE-EXPRESSION ,Genome ,Sequence Analysis, RNA ,ved/biology ,RNA ,Vertebrate ,PROFILES ,PATTERNS ,RNA, Long Noncoding - Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. A deeper characterization of regional transcription patterns within different heart chambers may aid to improve our understanding of the molecular mechanisms involved in myocardial function and further, our ability to develop novel therapeutic strategies. Here, we used RNA sequencing to determine differentially expressed protein coding (PC) and long non-coding (lncRNA) transcripts within the heart chambers across seven vertebrate species and identified evolutionarily conserved chamber specific genes, lncRNAs and pathways. We investigated lncRNA homologs based on sequence, secondary structure, synteny and expressional conservation and found most lncRNAs to be conserved by synteny. Regional co-expression patterns of transcripts are modulated by multiple factors, including genomic overlap, strandedness and transcript biotype. Finally, we provide a community resource designated EvoACTG, which informs researchers on the conserved yet intertwined nature of the coding and non-coding cardiac transcriptome across popular model organisms in CVD research.
- Published
- 2021
- Full Text
- View/download PDF
20. Grand challenges in molecular cardiology
- Author
-
Leon J. De Windt
- Published
- 2022
- Full Text
- View/download PDF
21. Dichotomy between the transcriptomic landscape of naturally versus accelerated aged murine hearts
- Author
-
Monika Stoll, Anne-Sophie Armand, Leon J. De Windt, Thomas Thum, Martine de Boer, Federica De Majo, Blanche Schroen, Jana-Charlotte Hegenbarth, Dirk J. Duncker, Frank Rühle, Christian Bär, RS: FSE DMG, Cardiologie, RS: Carim - H05 Gene regulation, RS: Carim - H02 Cardiomyopathy, Biochemie, RS: FHML MaCSBio, RS: Carim - B01 Blood proteins & engineering, Cardiology, Maastricht University [Maastricht], Institute of Molecular Biology (IMB), Johannes Gutenberg - Universität Mainz (JGU), University Hospital Münster - Universitaetsklinikum Muenster [Germany] (UKM), Hannover Medical School [Hannover] (MHH), Erasmus University Medical Center [Rotterdam] (Erasmus MC), Institut Necker Enfants-Malades (INEM - UM 111 (UMR 8253 / U1151)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), T.T. was supported by ERC Consolidator Grant LONGHEART, by ERA-CVD JCT2016 EXPERT and the DFG (TH903/22-1). C.B. was supported by the DFG (BA5631/2–1) and ERA-CVD JCT2016 EXPERT. D.J.D. acknowledges support from the Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation, Dutch Federation of University Medical Centers, ZonMW and the Royal Netherlands Academy of Sciences (CVON2011-ARENA and CVON2017-ARENA PRIME). A.S.A. was funded by Association Française contres les Myopathies (AFM 18802) L.D.W. and F.D.M. are supported by ERA-CVD JCT2016 EXPERT. L.D.W. acknowledges support from the Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation, Dutch Federation of University Medical Centers, ZonMW and the Royal Netherlands Academy of Sciences (CVON2017-ARENA PRIME). L.D.W. was further supported by ERC Consolidator Grant 311549 CALMIRS and a VICI award 918-156-47 from NWO. B.S. is supported by VIDI award 917.14.363 from NWO, Dekker grant 2014T105 from the Dutch Heart Foundation, further support comes from the Netherlands CardioVascular Research Initiative: the Dutch Heart Foundation, Dutch Federation of University Medical Centers, ZonMW and the Royal Netherlands Academy of Sciences (CVON2018 SHE-PREDICTS-HF), and a grant by the Health Foundation Limburg., Johannes Gutenberg - Universität Mainz = Johannes Gutenberg University (JGU), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), and Bodescot, Myriam
- Subjects
0301 basic medicine ,Male ,Aging ,lcsh:Medicine ,030204 cardiovascular system & hematology ,Gene regulatory networks ,Transcriptome ,Mice ,0302 clinical medicine ,FAILURE ,LONGEVITY ,lcsh:Science ,MUTATION ,Telomere Shortening ,Multidisciplinary ,Aging, Premature ,Heart ,Telomere ,CANCER ,Cell biology ,Mitochondria ,PREDICTS ,Antioxidant capacity ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Female ,NUCLEAR ABNORMALITIES ,Senescence ,Premature aging ,EXPRESSION ,DNA repair ,Biology ,Article ,Cardiac dysfunction ,MECHANISMS ,03 medical and health sciences ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Animals ,Humans ,Transcriptomics ,Gene ,Myocardium ,lcsh:R ,Proteins ,030104 developmental biology ,DNA-DAMAGE ,TELOMERE LENGTH ,lcsh:Q ,Ichthyosis, Lamellar - Abstract
We investigated the transcriptomic landscape of the murine myocardium along the course of natural aging and in three distinct mouse models of premature aging with established aging-related cardiac dysfunction. Genome-wide total RNA-seq was performed and the expression patterns of protein-coding genes and non-coding RNAs were compared between hearts from naturally aging mice, mice with cardiac-specific deficiency of a component of the DNA repair machinery, mice with reduced mitochondrial antioxidant capacity and mice with reduced telomere length. Our results demonstrate that no dramatic changes are evident in the transcriptomes of naturally senescent murine hearts until two years of age, in contrast to the transcriptome of accelerated aged mice. Additionally, these mice displayed model-specific alterations of the expression levels of protein-coding and non-coding genes with hardly any overlap with age-related signatures. Our data demonstrate very limited similarities between the transcriptomes of all our murine aging models and question their reliability to study human cardiovascular senescence.
- Published
- 2020
- Full Text
- View/download PDF
22. Circulating miR-216a as a biomarker of metabolic alterations and obesity in women
- Author
-
Zenab Mohseni, Chahinda Ghossein-Doha, Indira G. C. Vonhögen, Thomas Thum, Martina Calore, Bjorn Winkens, Ke Xiao, Marc E. A. Spaanderman, Paula A. da Costa Martins, Leon J. De Windt, RS: FSE DMG, Molecular Genetics, RS: Carim - H05 Gene regulation, Cardiologie, Obstetrie & Gynaecologie, FHML Methodologie & Statistiek, RS: CAPHRI - R6 - Promoting Health & Personalised Care, MUMC+: MA Medische Staf Obstetrie Gynaecologie (9), and RS: GROW - R4 - Reproductive and Perinatal Medicine
- Subjects
0301 basic medicine ,medicine.medical_specialty ,SAMPLE ,lcsh:QH426-470 ,MICRORNAS ,Vascular damage Radboud Institute for Health Sciences [Radboudumc 16] ,Type 2 diabetes ,PROFILE ,Biochemistry ,Article ,DISEASE ,SERUM ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,High-density lipoprotein ,Internal medicine ,Genetics ,medicine ,SYSTEMATIC ANALYSIS ,Obesity ,Molecular Biology ,GENE-EXPRESSION ,RISK ,Predictive marker ,medicine.diagnostic_test ,microRNA ,business.industry ,Biochemistry (medical) ,Fatty liver ,Biomarker ,GLOBAL BURDEN ,medicine.disease ,Metabolic syndrome ,lcsh:Genetics ,030104 developmental biology ,Endocrinology ,chemistry ,MIRNAS ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,COMPARATIVE RISK-ASSESSMENT ,Lipid profile ,business ,CLUSTERS ,Body mass index - Abstract
Contains fulltext : 229383.pdf (Publisher’s version ) (Open Access) Obesity leads to an amplified risk of disease and contributes to the occurrence of type 2 diabetes, fatty liver disease, coronary heart disease, stroke, chronic kidney disease and various types of cancer. MicroRNAs (miRNAs), small non-coding RNA molecules of 20-25 nucleotides, can remain stable in plasma and have been studied as potential (predictive) biomarkers for obesity and related metabolic disorders. The aim of this study was to identify circulating miRNAs as biomarkers for obesity status and metabolic alterations in women. Circulating miR-216a and miR-155-5p were selected by miRNA expression profiling and validated by real time quantitative PCR in a validation cohort of 60 obese women and 60 normal weight-age-matched control women. This was supplemented by correlation analysis of the candidate miRNA and anthropometric variables, blood biochemistry and lipid profile markers. Circulating miR-216a was validated as a biomarker of obesity status with significantly reduced levels in obese women. Interestingly, this was associated with a negative correlation between the plasma miR-216a content and body mass index (BMI), waist circumference, mean arterial pressure (MAP), triglycerides, ratio of total cholesterol/high density lipoprotein (HDL)-cholesterol and high sensitivity-C reactive protein (hs-CRP).Taken together, we provide evidence for an abnormally expressed circulating miRNA, miR-216a, with additive value as a predictive marker for obesity that correlates with metabolic alterations presented by lipid profile and inflammatory markers.
- Published
- 2020
- Full Text
- View/download PDF
23. Intercellular transfer of miR-200c-3p impairs the angiogenic capacity of cardiac endothelial cells
- Author
-
Lara Ottaviani, Rio P. Juni, Ricardo C. de Abreu, Marida Sansonetti, Vasco Sampaio-Pinto, Julie Halkein, Jana C. Hegenbarth, Nadja Ring, Kevin Knoops, Jordy M.M. Kocken, Carlos de Jesus, Auriane C. Ernault, Hamid el Azzouzi, Frank Rühle, Servé Olieslagers, Hugo Fernandes, Lino Ferreira, Luca Braga, Monika Stoll, Diana S. Nascimento, Leon J. de Windt, Paula A. da Costa Martins, Physiology, ACS - Heart failure & arrhythmias, ACS - Microcirculation, Molecular Genetics, RS: Carim - H05 Gene regulation, Cardiologie, RS: FSE DMG, RS: M4I - Nanoscopy, Institute of Nanoscopy (IoN), Microscopy CORE Lab, Biochemie, and RS: Carim - B01 Blood proteins & engineering
- Subjects
EXOSOMES ,Pharmacology ,PROLIFERATION ,Endothelial Cells ,Cell Communication ,CARDIOMYOCYTES ,APOPTOSIS ,ACTIVATION ,HYPERTROPHY ,Extracellular Vesicles ,Mice ,MicroRNAs ,Drug Discovery ,Genetics ,FAILURE ,HEART ,Molecular Medicine ,Animals ,Original Article ,Myocytes, Cardiac ,MESSENGER-RNAS ,Molecular Biology - Abstract
As mediators of intercellular communication, extracellular vesicles containing molecular cargo, such as microRNAs, are secreted by cells and taken up by recipient cells to influence their cellular phenotype and function. Here we report that cardiac stress-induced differential microRNA content, with miR-200c-3p being one of the most enriched, in cardiomyocyte-derived extracellular vesicles mediates functional cross-talk with endothelial cells. Silencing of miR-200c-3p in mice subjected to chronic increased cardiac pressure overload resulted in attenuated hypertrophy, smaller fibrotic areas, higher capillary density, and preserved cardiac ejection fraction. We were able to maximally rescue microvascular and cardiac function with very low doses of antagomir, which specifically silences miR-200c-3p expression in non-myocyte cells. Our results reveal vesicle transfer of miR-200c-3p from cardiomyocytes to cardiac endothelial cells, underlining the importance of cardiac intercellular communication in the pathophysiology of heart failure.
- Published
- 2022
- Full Text
- View/download PDF
24. Long Non-Coding RNAs in Cardiac Hypertrophy
- Author
-
Nicolò Mangraviti and Leon J. De Windt
- Abstract
Heart disease represents one of the main challenges in modern medicine with insufficient treatment options. Whole genome sequencing allowed for the discovery of several classes of non-coding RNA (ncRNA) and widened our understanding of disease regulatory circuits. The intrinsic ability of long ncRNAs (lncRNAs) and circular RNAs (circRNAs) to regulate gene expression by a plethora of mechanisms make them candidates for conceptually new treatment options. However, important questions remain to be addressed before we can fully exploit the therapeutic potential of these molecules. Increasing our knowledge of their mechanisms of action and refining the approaches for modulating lncRNAs expression are just a few of the challenges we face. The accurate identification of novel lncRNAs is hampered by their relatively poor cross-species sequence conservation and their low and context-dependent expression pattern. Nevertheless, progress has been made in their annotation in recent years, while a few experimental studies have confirmed the value of lncRNAs as new mechanisms in the development of cardiac hypertrophy and other cardiovascular diseases. Here, we explore cardiac lncRNA biology and the evidence that this class of molecules has therapeutic benefit to treat cardiac hypertrophy.
- Published
- 2022
- Full Text
- View/download PDF
25. Genomic instability in the naturally and prematurely aged myocardium
- Author
-
Martine de Boer, Frank Rühle, Vicente Andrés, Leon J. De Windt, Erika Hilbold, Christian Bär, Jana-Charlotte Hegenbarth, Monika Stoll, Rosa M Nevado, Blanche Schroen, Leonie Martens, Dirk J. Duncker, Anne-Sophie Armand, Federica De Majo, Thomas Thum, Magda R. Hamczyk, RS: FSE DMG, RS: Carim - H05 Gene regulation, Cardiologie, RS: Carim - H01 Clinical atrial fibrillation, RS: Carim - H02 Cardiomyopathy, Biochemie, RS: FHML MaCSBio, RS: Carim - B01 Blood proteins & engineering, Publica, Maastricht University (Países Bajos), CVON-ARENA-PRIME, University of Münster (Alemania), Ministerio de Ciencia e Innovación (España), Ministerio de Educación, Cultura y Deporte (España), Instituto de Salud Carlos III, ERA-CVD JCT2016 EXPERT Network, Unión Europea. Comisión Europea. H2020, Unión Europea. Fondo Europeo de Desarrollo Regional (FEDER/ERDF), Fundación ProCNIC, Ministerio de Ciencia e Innovación. Centro de Excelencia Severo Ochoa (España), Deutsche Forschungsgemeinschaft (Alemania), European Research Council, Dutch CardioVascular Initiative, Netherlands Heart Foundation, Dutch Federation of University Medical Centers, Association Française contres les Myopathies, Marie Curie, and Cardiology
- Subjects
Genome instability ,Male ,Aging ,DNA Repair ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Mice ,0302 clinical medicine ,LONGEVITY ,MUTATION ,Cellular Senescence ,0303 health sciences ,Mutation ,Progeria ,Multidisciplinary ,DEATH ,Aging, Premature ,Heart ,Biological Sciences ,aging  ,genomic instability  ,CANCER ,Cell biology ,Mitochondria ,DNA-Binding Proteins ,Knockout mouse ,Female ,NUCLEAR ABNORMALITIES ,Premature aging ,EXPRESSION ,DNA repair ,DNA damage ,Biology ,Genomic Instability ,03 medical and health sciences ,medicine ,Animals ,030304 developmental biology ,DNA repair  ,Myocardium ,oxidative stress  ,medicine.disease ,Endonucleases ,FRAMEWORK ,Telomere ,Mice, Inbred C57BL ,Disease Models, Animal ,MICE ,DNA-DAMAGE ,TELOMERE LENGTH ,RNA-seq ,DNA Damage - Abstract
Genomic instability, the unresolved accumulation of DNA variants, is hypothesized as one of the contributors to the natural aging process. We assessed the frequency of unresolved DNA damage reaching the transcriptome of the murine myocardium during the course of natural aging and in hearts from four distinct mouse models of premature aging with established aging-related cardiac dysfunctions. RNA sequencing and variant calling based on total RNA sequencing was compared between hearts from naturally aging mice, mice with cardiomyocyte-specific deficiency of Ercc1, a component of the DNA repair machinery, mice with reduced mitochondrial antioxidant capacity, Tert-deficient mice with reduced telomere length, and a mouse model of human Hutchinson-Gilford progeria syndrome (HGPS). Our results demonstrate that no enrichment in variants is evident in the naturally aging murine hearts until 2 y of age from the HGPS mouse model or mice with reduced telomere lengths. In contrast, a dramatic accumulation of variants was evident in Ercc1 cardiomyocyte-specific knockout mice with deficient DNA repair machinery, in mice with reduced mitochondrial antioxidant capacity, and in the intestine, liver, and lung of naturally aging mice. Our data demonstrate that genomic instability does not evidently contribute to naturally aging of the mouse heart in contrast to other organs and support the contention that the endogenous DNA repair machinery is remarkably active to maintain genomic integrity in cardiac cells throughout life. F.D.M. is supported by HS-BAFTA and Kootstra fellowships of Maastricht University and a CVON-ARENA-PRIME fellowship. L.M. is supported by the fund Innovative Medical Research of the University of Münster Medical School (RÜ121510). M.R.H. is supported by a Juan de la Cierva contract from the Spanish Ministerio de Ciencia, Innovación y Universidades (IJC2019-040798-I). R.M.N. is the beneficiary of a predoctoral contract from the Spanish Ministerio de Educación, Cultura y Deporte (FPU16/ 05027). V.A. is supported by the Spanish Ministerio de Ciencia e Innovación (PID2019-108489RB-I00) and the Instituto de Salud Carlos III (ISCIII) (AC16/ 00091) as member of the ERA-CVD JCT2016 EXPERT Network (European Union’s Horizon 2020 Framework Programme), with cofunding from the European Regional Development Fund (“Una manera de hacer Europa”). The Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) is supported by the Ministry for Research, Science and Innovation (MICIN), the ISCIII, the Pro-CNIC Foundation, and is a Severo Ochoa Center of Excellence. C.B. was supported by the Deutsche Forschungsgemeinschaft (DFG) (BA5631/ 2-1). T.T. was supported by the European Research Council (ERC) Consolidator Grant LONGHEART, by ERA-CVD JCT2016 EXPERT, and the DFG (TH903/ 22-1). D.J.D., M.S., and L.J.D.W. acknowledge support from the Dutch CardioVascular Initiative: the Netherlands Heart Foundation, Dutch Federation of University Medical Centers, ZonMW, and the Royal Netherlands Academy of Sciences (CVON-ARENA-PRIME, CVON-RACE-V, CVON-PREDICT-2). B.S. acknowledges funding by the Netherlands Heart Foundation (Dr. Dekker 2014T105 and CVON-SHE-PREDICTS-HF) and a VIDI Award 917.14.363 from the Dutch Research Council (NWO). A.S.A. was funded by Association Française contres les Myopathies (AFM 18802). F.D.M., T.T., and L.J.D.W. are supported by ERA-CVD JCT2016 EXPERT. M.S. is funded by the DFG (RTG2220, Project 281125614) and Marie Skłodowska-Curie Grant Agreement 81371. L.J.D.W. was further supported by ERC Consolidator Grant 311549 CALMIRS, a VICI Award 918-156-47 from NWO and Marie Skłodowska-Curie Grant Agreements 813716 and 765274. Sí
- Published
- 2021
26. A microRNA program regulates the balance between cardiomyocyte hyperplasia and hypertrophy and stimulates cardiac regeneration
- Author
-
Vasco Sampaio-Pinto, Lara Ottaviani, Luca Braga, Roel A. de Weger, Leon J. De Windt, Lorena Zentillin, Daniel W. Sorensen, Jop H. van Berlo, Andrea Raso, Serena Zacchigna, Silvia Moimas, Paula A. da Costa Martins, Diana S. Nascimento, Manon M. H. Huibers, Hamid el Azzouzi, Ellen Dirkx, Sailay Siddiqi, Ryan John Cubero, Servé Olieslagers, Consuelo Torrini, Mauro Giacca, Raso, A., Dirkx, E., Sampaio-Pinto, V., el Azzouzi, H., Cubero, R. J., Sorensen, D. W., Ottaviani, L., Olieslagers, S., Huibers, M. M., de Weger, R., Siddiqi, S., Moimas, S., Torrini, C., Zentillin, L., Braga, L., Nascimento, D. S., da Costa Martins, P. A., van Berlo, J. H., Zacchigna, S., Giacca, M., De Windt, L. J., RS: Carim - Heart, Cardiologie, RS: FSE DMG, RS: Carim - H05 Gene regulation, and Molecular Genetics
- Subjects
Myocardial Infarction ,General Physics and Astronomy ,Muscle hypertrophy ,ACTIVATION ,PATHWAY ,Mice ,Myocyte ,Myocytes, Cardiac ,Cells, Cultured ,IN-VIVO ,Regulation of gene expression ,Multidisciplinary ,Cultured ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,PROLIFERATION ,food and beverages ,MicroRNA ,Hyperplasia ,Cell cycle ,Cell biology ,Echocardiography ,miRNAs ,Cardiac regeneration ,HAND2 ,Cardiac ,Human ,HEART REGENERATION ,EXPRESSION ,Science ,Cells ,Cardiomegaly ,Gene delivery ,MYOCYTES ,Article ,General Biochemistry, Genetics and Molecular Biology ,ADULT ,REGRESSION ,medicine ,Animals ,Humans ,Regeneration ,Animals, Newborn ,Gene Expression Regulation ,MicroRNAs ,Rats ,Animal ,Regeneration (biology) ,General Chemistry ,medicine.disease ,Newborn ,ATRIAL-FIBRILLATION ,biology.protein ,Rat - Abstract
Myocardial regeneration is restricted to early postnatal life, when mammalian cardiomyocytes still retain the ability to proliferate. The molecular cues that induce cell cycle arrest of neonatal cardiomyocytes towards terminally differentiated adult heart muscle cells remain obscure. Here we report that the miR-106b~25 cluster is higher expressed in the early postnatal myocardium and decreases in expression towards adulthood, especially under conditions of overload, and orchestrates the transition of cardiomyocyte hyperplasia towards cell cycle arrest and hypertrophy by virtue of its targetome. In line, gene delivery of miR-106b~25 to the mouse heart provokes cardiomyocyte proliferation by targeting a network of negative cell cycle regulators including E2f5, Cdkn1c, Ccne1 and Wee1. Conversely, gene-targeted miR-106b~25 null mice display spontaneous hypertrophic remodeling and exaggerated remodeling to overload by derepression of the prohypertrophic transcription factors Hand2 and Mef2d. Taking advantage of the regulatory function of miR-106b~25 on cardiomyocyte hyperplasia and hypertrophy, viral gene delivery of miR-106b~25 provokes nearly complete regeneration of the adult myocardium after ischemic injury. Our data demonstrate that exploitation of conserved molecular programs can enhance the regenerative capacity of the injured heart., Myocardial regeneration and proliferation of heart muscle cells is limited to a short period after birth early postnatal life, after which heart muscle cells can only grow in size and not in number. Here, the authors identified that the expression level of an endogenous microRNA cluster in heart muscle promotes the passage of the proliferative state to adult heart growth, and modulating the expression of this cluster can stimulate heart regeneration after myocardial infarction.
- Published
- 2021
- Full Text
- View/download PDF
27. A novel murine model for arrhythmogenic cardiomyopathy points to a pathogenic role of Wnt signalling and miRNA dysregulation
- Author
-
Mohsin A.F. Khan, Giuseppe Lembo, Paolo Bonaldo, Daniela Carnevale, Alessandra Lorenzon, Alessandra Rampazzo, Patrizia Sabatelli, Claudia Sacchetto, Roberto Doliana, Libero Vitiello, Leon J. De Windt, Roman S. Polishchuk, Giorgia Beffagna, Paola Braghetta, Giulia Poloni, Martina Calore, Emanuela Dazzo, Cardiologie, RS: FSE DMG, RS: CARIM - R2 - Cardiac function and failure, and RS: Carim - H05 Gene regulation
- Subjects
0301 basic medicine ,NUCLEAR PLAKOGLOBIN ,Physiology ,Arrhythmogenic cardiomyopathy ,Cardiomyopathy ,030204 cardiovascular system & hematology ,medicine.disease_cause ,CARDIOMYOCYTES ,Pathogenesis ,0302 clinical medicine ,Gene Regulatory Networks ,INTERCALATED DISKS ,Wnt Signaling Pathway ,Arrhythmogenic Right Ventricular Dysplasia ,Mutation ,Desmoglein 2 ,Arrhythmogenic cardiomyopathy Molecular pathogenesis miRNA ,biology ,Wnt signaling pathway ,PROLIFERATION ,Phenotype ,LEADS ,Codon, Nonsense ,Cardiology and Cardiovascular Medicine ,STEM-CELLS ,RIGHT-VENTRICULAR CARDIOMYOPATHY ,Nonsense mutation ,Molecular pathogenesis ,miRNA ,CARDIAC DIFFERENTIATION ,Mice, Transgenic ,Sudden death ,CONTRIBUTES ,03 medical and health sciences ,Physiology (medical) ,medicine ,Animals ,Genetic Predisposition to Disease ,Desmoplakin ,Myocardium ,Editorials ,medicine.disease ,Fibrosis ,ADIPOGENESIS ,Mice, Inbred C57BL ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,Gene Expression Regulation ,biology.protein ,Cancer research ,Transcriptome - Abstract
Aims Arrhythmogenic cardiomyopathy (AC) is one of the most common inherited cardiomyopathies, characterized by progressive fibro-fatty replacement in the myocardium. Clinically, AC manifests itself with ventricular arrhythmias, syncope, and sudden death and shows wide inter- and intra-familial variability. Among the causative genes identified so far, those encoding for the desmosomal proteins plakophilin-2 (PKP2), desmoplakin (DSP), and desmoglein-2 (DSG2) are the most commonly mutated. So far, little is known about the molecular mechanism(s) behind such a varied spectrum of phenotypes, although it has been shown that the causative mutations not only lead to structural abnormalities but also affect the miRNA profiling of cardiac tissue. Here, we aimed at studying the pathogenic effects of a nonsense mutation of the desmoglein-2 gene, both at the structural level and in terms of miRNA expression pattern.Methods and results We generated transgenic mice with cardiomyocyte-specific overexpression of a FLAG-tagged human desmoglein-2 harbouring the Q558* nonsense mutation found in an AC patient. The hearts of these mice showed signs of fibrosis, decrease in desmosomal size and number, and reduction of the Wnt/beta-catenin signalling. Genome-wide RNA-Seq performed in Tg-hQ hearts and non-transgenic hearts revealed that 24 miRNAs were dysregulated in transgenic animals. Further bioinformatic analyses for selected miRNAs suggested that miR-217-5p, miR-499-5p, and miR-708-5p might be involved in the pathogenesis of the disease.Conclusion Down-regulation of the canonical Wnt/beta-catenin signalling might be considered a common key event in the AC pathogenesis. We identified the miRNA signature in AC hearts, with miR-708-5p and miR-217-5p being the most up-regulated and miR-499-5p the most down-regulated miRNAs. All of them were predicted to be involved in the regulation of the Wnt/beta-catenin pathway and might reveal the potential pathophysiology mechanisms of AC, as well as be useful as therapeutic targets for the disease.
- Published
- 2019
- Full Text
- View/download PDF
28. The continuous heart failure spectrum: moving beyond an ejection fraction classification
- Author
-
Gilles W. De Keulenaer, Robert O. Bonow, Nazha Hamdani, Filippos Triposkiadis, Johann Bauersachs, Richard T. Lee, Vincent F.M. Segers, Ida G. Lunde, Douglas L. Mann, John Parissis, Petros Nihoyannopoulos, Johannes Backs, Alexandre Mebazaa, Vijay K. Chopra, Stephane Heymans, Randall C. Starling, Javed Butler, Ajay M. Shah, Giuseppe M.C. Rosano, Christoph Maack, Alexander R. Lyon, Jean L. Rouleau, Jean Noel Trochu, Wolfgang A. Linke, Zoltán Papp, Rudolf A. de Boer, Marvin A. Konstam, Robert J. Mentz, Thomas Thum, Francois M. Abboud, Petar M. Seferović, Jean-Sébastien Hulot, Carlo G. Tocchetti, Stamatis Adamopoulos, Dirk L. Brutsaert, Faiez Zannad, Gerd Hasenfuss, John Atherton, Leon J. De Windt, Daniel Burkhoff, Paul W. Armstrong, Thierry Pedrazzini, CIC - HEGP (CIC 1418), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Paris-Centre de Recherche Cardiovasculaire (PARCC (UMR_S 970/ U970)), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), Triposkiadis, Filippo, Butler, Javed, Abboud, Francois M, Armstrong, Paul W, Adamopoulos, Stamati, Atherton, John J, Backs, Johanne, Bauersachs, Johann, Burkhoff, Daniel, Bonow, Robert O, Chopra, Vijay K, de Boer, Rudolf A, de Windt, Leon, Hamdani, Nazha, Hasenfuss, Gerd, Heymans, Stephane, Hulot, Jean-Sébastien, Konstam, Marvin, Lee, Richard T, Linke, Wolfgang A, Lunde, Ida G, Lyon, Alexander R, Maack, Christoph, Mann, Douglas L, Mebazaa, Alexandre, Mentz, Robert J, Nihoyannopoulos, Petro, Papp, Zoltan, Parissis, John, Pedrazzini, Thierry, Rosano, Giuseppe, Rouleau, Jean, Seferovic, Petar M, Shah, Ajay M, Starling, Randall C, Tocchetti, Carlo G, Trochu, Jean-Noel, Thum, Thoma, Zannad, Faiez, Brutsaert, Dirk L, Segers, Vincent F, and De Keulenaer, Gilles W
- Subjects
Clinical Review ,Ejection fraction ,Cardiac & Cardiovascular Systems ,[SDV]Life Sciences [q-bio] ,Disease ,Comorbidity ,030204 cardiovascular system & hematology ,EXERCISE CAPACITY ,law.invention ,Ventricular Dysfunction, Left ,0302 clinical medicine ,Randomized controlled trial ,law ,Reference Values ,Medicine ,Myocytes, Cardiac ,030212 general & internal medicine ,1102 Cardiorespiratory Medicine and Haematology ,OUTCOMES ,Ventricular Remodeling ,Stroke volume ,ASSOCIATION ,3. Good health ,PREVALENCE ,VENTRICULAR-FUNCTION ,Cardiology ,Disease Progression ,cardiovascular system ,Cardiology and Cardiovascular Medicine ,ECHOCARDIOGRAPHY ,Life Sciences & Biomedicine ,circulatory and respiratory physiology ,medicine.medical_specialty ,Heart failure ,Pathophysiology ,GLOBAL LONGITUDINAL STRAIN ,03 medical and health sciences ,Internal medicine ,CO-MORBIDITIES ,Humans ,cardiovascular diseases ,Endothelium ,Ventricular remodeling ,Science & Technology ,business.industry ,Stroke Volume ,medicine.disease ,DYSFUNCTION ,COMORBIDITIES ,Cardiovascular System & Hematology ,Cardiovascular System & Cardiology ,Human medicine ,Endothelium, Vascular ,business ,Heart failure with preserved ejection fraction - Abstract
Randomized clinical trials initially used heart failure (HF) patients with low left ventricular ejection fraction (LVEF) to select study populations with high risk to enhance statistical power. However, this use of LVEF in clinical trials has led to oversimplification of the scientific view of a complex syndrome. Descriptive terms such as ‘HFrEF’ (HF with reduced LVEF), ‘HFpEF’ (HF with preserved LVEF), and more recently ‘HFmrEF’ (HF with mid-range LVEF), assigned on arbitrary LVEF cut-off points, have gradually arisen as separate diseases, implying distinct pathophysiologies. In this article, based on pathophysiological reasoning, we challenge the paradigm of classifying HF according to LVEF. Instead, we propose that HF is a heterogeneous syndrome in which disease progression is associated with a dynamic evolution of functional and structural changes leading to unique disease trajectories creating a spectrum of phenotypes with overlapping and distinct characteristics. Moreover, we argue that by recognizing the spectral nature of the disease a novel stratification will arise from new technologies and scientific insights that will shape the design of future trials based on deeper understanding beyond the LVEF construct alone.
- Published
- 2021
- Full Text
- View/download PDF
29. Contributors
- Author
-
Bence Ágg, Parisa Aghagolzadeh, Chukwuemeka George Anene-Nzelu, Johannes Backs, Ferran Barbé, Fay Betsou, Stephanie Bezzina Wettinger, Andrei Codreanu, Yvan Devaux, Christoph Dieterich, Javier Durán, Rosienne Farrugia, Kyriacos Felekkis, Péter Ferdinandy, Roger S-Y Foo, Eleftheria Galatou, David de Gonzalo-Calvo, Simona Greco, Johannes Grillari, Hakan Gunes, Matthias Hackl, Nazha Hamdani, Lutz Hein, Carlos Hermenegildo, Eduardo Iglesias-Gutiérrez, Benedetta Izzi, Kornelia Jaquet, Amela Jusic, Kanita Karađuzović-Hadžiabdić, Gabriela M. Kuster, Alisia Madè, Federica De Majo, Fabio Martelli, Andreas Mügge, Vivien Ngo, Susana Novella, Ana Belén Paes, Christos Papaneophytou, Thierry Pedrazzini, Antje Peters, Lucía Pinilla, Emma Louise Robinson, Elisabeth Semmelrock, Justus Stenzig, Maarten Vanhaverbeke, Mirko Völkers, Leon J. De Windt, Johannes Winkler, Angela Xuereb Anastasi, and Mehmet Birhan Yilmaz
- Published
- 2021
- Full Text
- View/download PDF
30. Circulating miR-185-5p as a Potential Biomarker for Arrhythmogenic Right Ventricular Cardiomyopathy
- Author
-
Riccardo Bariani, Domenico Corrado, Luciano Daliento, Ke Xiao, Indira G. C. Vonhögen, Chiara Romualdi, Barbara Bauce, Libero Vitiello, Leon J. De Windt, Alessandra Rampazzo, Marzia De Bortoli, Alessandra Lorenzon, Thomas Thum, Giulia Poloni, Robin M. W. Colpaert, Martina Calore, Elisa Mazzotti, Claudia Sacchetto, Zenab Mohseni, Publica, Molecular Genetics, RS: Carim - H05 Gene regulation, Cardiologie, and RS: FSE DMG
- Subjects
Adult ,Male ,medicine.medical_specialty ,QH301-705.5 ,heart failure ,Pilot Projects ,Disease ,030204 cardiovascular system & hematology ,Sudden death ,Article ,Right ventricular cardiomyopathy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,microRNA ,medicine ,Humans ,genetics ,Biology (General) ,Arrhythmogenic Right Ventricular Dysplasia ,030304 developmental biology ,arrhythmogenic right ventricular cardiomyopathy ,0303 health sciences ,business.industry ,MICRORNA ,Area under the curve ,biomarkers ,General Medicine ,medicine.disease ,Pathophysiology ,Circulating MicroRNA ,MicroRNAs ,Case-Control Studies ,Heart failure ,Cardiology ,circulating microRNAs ,Female ,Cardiomyopathies ,business - Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a genetic cardiac disease characterized by progressive myocardial fibro-fatty replacement, arrhythmias and risk of sudden death. Its diagnosis is challenging and often it is achieved after disease onset or postmortem. In this study, we sought to identify circulating microRNAs (miRNAs) differentially expressed in ARVC patients compared to healthy controls. In the pilot study, we screened the expression of 754 miRNAs from 21 ARVC patients and 20 healthy controls. After filtering the miRNAs considering a log fold-change cut-off of ±1, p-value <, 0.05, we selected five candidate miRNAs for a subsequent validation study in which we used TaqMan-based real-time PCR to analyse samples from 37 ARVC patients and 30 healthy controls. We found miR-185-5p significantly upregulated in ARVC patients. Receiver operating characteristic analysis indicated an area under the curve of 0.854, corroborating the link of this miRNA and ARVC pathophysiology.
- Published
- 2021
31. Non-coding RNAs in cardiac inflammation: key drivers in the pathophysiology of heart failure
- Author
-
Marida Sansonetti, Leon J. De Windt, Cardiologie, RS: Carim - H05 Gene regulation, and RS: FSE DMG
- Subjects
0301 basic medicine ,EXPRESSION ,Physiology ,Inflammatory response ,Coding (therapy) ,Inflammation ,MACROPHAGE ACTIVATION ,030204 cardiovascular system & hematology ,Bioinformatics ,DENDRITIC CELLS ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,IMMUNE CELL ,Physiology (medical) ,medicine ,Humans ,CD4(+) T-LYMPHOCYTES ,Heart Failure ,CUTTING EDGE ,business.industry ,MONOCYTE CHEMOATTRACTANT PROTEIN-1 ,ncRNAs ,Arrhythmias, Cardiac ,Heart ,medicine.disease ,TNF-ALPHA ,Pathophysiology ,Inflammatory cells ,MicroRNAs ,030104 developmental biology ,Cardiac inflammation ,MYOCARDIAL-INFARCTION ,Heart failure ,MAST-CELLS ,RNA, Long Noncoding ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Abstract
Heart failure is among the most progressive diseases and a leading cause of morbidity. Despite several advances in cardiovascular therapies, pharmacological treatments are limited to relieve symptoms without curing cardiac injury. Multiple observations point to the involvement of immune cells as key drivers in the pathophysiology of heart failure. In particular, there is a growing recognition that heart failure is related to a prolonged and insufficiently repressed inflammatory response leading to molecular, cellular, and functional cardiac alterations. Over the last decades, non-coding RNAs are recognized as prominent mediators of cardiac inflammation, affecting the function of several immune cells. In the current review, we explore the contribution of the diverse immune cells in the progression of heart failure, revealing mechanistic functions for non-coding RNAs in cardiac immune cells as a new and exciting field of investigation.
- Published
- 2020
32. Modeling Cardiovascular Diseases with hiPSC-Derived Cardiomyocytes in 2D and 3D Cultures
- Author
-
Alessandra Rampazzo, Martina Calore, Libero Vitiello, Leon J. De Windt, and Claudia Sacchetto
- Subjects
0301 basic medicine ,Cardiomyopathy ,Cell Culture Techniques ,improve recovery ,Review ,030204 cardiovascular system & hematology ,cardiac disease modeling ,functional maturation ,Extracellular matrix ,lcsh:Chemistry ,0302 clinical medicine ,3D cardiac models ,Myocytes, Cardiac ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Spectroscopy ,Cells, Cultured ,Tissue Scaffolds ,pluripotent stem-cell ,Models, Cardiovascular ,METABOLIC MATURATION ,Cell Differentiation ,General Medicine ,Computer Science Applications ,Cell biology ,Drug development ,Cardiovascular Diseases ,engineered heart tissue ,human induced pluripotent stem cells ,engineered human myocardium ,Cell type ,Induced Pluripotent Stem Cells ,Biology ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,medicine ,Humans ,Physical and Theoretical Chemistry ,Human Induced Pluripotent Stem Cells ,Molecular Biology ,QT SYNDROME ,CARDIOMYOPATHY ,Tissue Engineering ,Organic Chemistry ,Disease mechanisms ,tissue ,medicine.disease ,ENDOTHELIAL-CELLS ,CARDIAC MICROTISSUES ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cell culture - Abstract
In the last decade, the generation of cardiac disease models based on human-induced pluripotent stem cells (hiPSCs) has become of common use, providing new opportunities to overcome the lack of appropriate cardiac models. Although much progress has been made toward the generation of hiPSC-derived cardiomyocytes (hiPS-CMs), several lines of evidence indicate that two-dimensional (2D) cell culturing presents significant limitations, including hiPS-CMs immaturity and the absence of interaction between different cell types and the extracellular matrix. More recently, new advances in bioengineering and co-culture systems have allowed the generation of three-dimensional (3D) constructs based on hiPSC-derived cells. Within these systems, biochemical and physical stimuli influence the maturation of hiPS-CMs, which can show structural and functional properties more similar to those present in adult cardiomyocytes. In this review, we describe the latest advances in 2D- and 3D-hiPSC technology for cardiac disease mechanisms investigation, drug development, and therapeutic studies.
- Published
- 2020
33. MiR-337-3p Promotes Adipocyte Browning by Inhibiting TWIST1
- Author
-
Mora Murri, Paula A. da Costa Martins, Indira G. C. Vonhögen, Francisco J. Tinahones, Servé Olieslagers, Hamid el Azzouzi, Aliaksei S Vasilevich, Leon J. De Windt, Jan de Boer, Biointerface Science, ICMS Core, EAISI Health, RS: FSE DMG, RS: Carim - H05 Gene regulation, Cardiologie, RS: MERLN - Cell Biology - Inspired Tissue Engineering (CBITE), CBITE, and Molecular Genetics
- Subjects
obesity ,Regulator ,Adipose tissue ,White adipose tissue ,Mitochondrion ,SDG 3 – Goede gezondheid en welzijn ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Adipose Tissue, Brown ,Adipocyte ,Brown adipose tissue ,lcsh:QH301-705.5 ,Feedback, Physiological ,Metabolic Syndrome ,0303 health sciences ,microRNA ,Nuclear Proteins ,Thermogenesis ,General Medicine ,Up-Regulation ,mitochondria ,medicine.anatomical_structure ,ADIPOSE-TISSUE ,Adipocytes, Brown ,030220 oncology & carcinogenesis ,medicine.medical_specialty ,MITOCHONDRIAL DYSFUNCTION ,animal structures ,Biology ,Article ,metabolic syndrome ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,Animals ,Humans ,030304 developmental biology ,Base Sequence ,Twist-Related Protein 1 ,brown adipose tissue ,medicine.disease ,MicroRNAs ,Endocrinology ,chemistry ,lcsh:Biology (General) ,FAT ,Metabolic syndrome - Abstract
The prevalence of metabolic syndrome (MetS) and obesity is an alarming health issue worldwide. Obesity is characterized by an excessive accumulation of white adipose tissue (WAT), and it is associated with diminished brown adipose tissue (BAT) activity. Twist1 acts as a negative feedback regulator of BAT metabolism. Therefore, targeting Twist1 could become a strategy for obesity and metabolic disease. Here, we have identified miR-337-3p as an upstream regulator of Twist1. Increased miR-337-3p expression paralleled decreased expression of TWIST1 in BAT compared to WAT. Overexpression of miR-337-3p in brown pre-adipocytes provoked a reduction in Twist1 expression that was accompanied by increased expression of brown/mitochondrial markers. Luciferase assays confirmed an interaction between the miR-337 seed sequence and Twist1 3&prime, UTR. The inverse relationship between the expression of TWIST1 and miR-337 was finally validated in adipose tissue samples from non-MetS and MetS subjects that demonstrated a dysregulation of the miR-337-Twist1 molecular axis in MetS. The present study demonstrates that adipocyte miR-337-3p suppresses Twist1 repression and enhances the browning of adipocytes.
- Published
- 2020
34. The MEF2 transcriptional target DMPK induces loss of sarcomere structure and cardiomyopathy
- Author
-
Abdelaziz Beqqali, Meriem Bourajjaj, Maarten M.G. van den Hoogenhof, Ingeborg van der Made, Benoit Samson-Couterie, Elisabeth Ehler, Amin Damanafshan, Leon J. De Windt, Yigal M. Pinto, Henk A. van Veen, Ies Elzenaar, Ralph J. van Oort, Daisy I. Picavet, RS: CARIM - R2.07 - Gene regulation, Cardiologie, Graduate School, ACS - Heart failure & arrhythmias, Cardiology, Cell Biology and Histology, and Medical Biology
- Subjects
Male ,0301 basic medicine ,Transcription, Genetic ,Physiology ,Cardiomyopathy ,030204 cardiovascular system & hematology ,Sarcomere ,Animals, Genetically Modified ,0302 clinical medicine ,Myocyte ,Myocytes, Cardiac ,SERUM RESPONSE FACTOR ,CARDIAC-HYPERTROPHY ,PHOSPHORYLATION ,GENE-EXPRESSION ,Ventricular Remodeling ,MEF2 Transcription Factors ,Cell biology ,HEART-FAILURE ,SRF ,MEF2 ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,Signal Transduction ,DMPK ,Sarcomeres ,Mef2 ,Gene isoform ,MYOTONIC-DYSTROPHY ,Heart failure ,Biology ,Myotonin-Protein Kinase ,SIGNALING PATHWAYS ,03 medical and health sciences ,Physiology (medical) ,Serum response factor ,medicine ,Animals ,Humans ,Rats, Wistar ,Enhancer ,Transcription factor ,DYSTROPHY PROTEIN-KINASE ,medicine.disease ,DILATED CARDIOMYOPATHY ,Mice, Inbred C57BL ,Disease Models, Animal ,MICE ,HEK293 Cells ,030104 developmental biology ,Animals, Newborn ,Transcription Factors - Abstract
AimsThe pathology of heart failure is characterized by poorly contracting and dilated ventricles. At the cellular level, this is associated with lengthening of individual cardiomyocytes and loss of sarcomeres. While it is known that the transcription factor myocyte enhancer factor-2 (MEF2) is involved in this cardiomyocyte remodelling, the underlying mechanism remains to be elucidated. Here, we aim to mechanistically link MEF2 target genes with loss of sarcomeres during cardiomyocyte remodelling.Methods and resultsNeonatal rat cardiomyocytes overexpressing MEF2 elongated and lost their sarcomeric structure. We identified myotonic dystrophy protein kinase (DMPK) as direct MEF2 target gene involved in this process. Adenoviral overexpression of DMPK E, the isoform upregulated in heart failure, resulted in severe loss of sarcomeres in vitro, and transgenic mice overexpressing DMPK E displayed disruption of sarcomere structure and cardiomyopathy in vivo. Moreover, we found a decreased expression of sarcomeric genes following DMPK E gain-of-function. These genes are targets of the transcription factor serum response factor (SRF) and we found that DMPK E acts as inhibitor of SRF transcriptional activity.ConclusionOur data indicate that MEF2-induced loss of sarcomeres is mediated by DMPK via a decrease in sarcomeric gene expression by interfering with SRF transcriptional activity. Together, these results demonstrate an unexpected role for DMPK as a direct mediator of adverse cardiomyocyte remodelling and heart failure.
- Published
- 2018
- Full Text
- View/download PDF
35. MicroRNA-221/222 Family Counteracts Myocardial Fibrosis in Pressure Overload-Induced Heart Failure
- Author
-
Mitchell Bijnen, Tessa van Herwaarden, Michiel T H M Henkens, Wouter Verhesen, Marie-José Goumans, Marc van Bilsen, Frans A. van Nieuwenhoven, Arantxa González, Tim J Peters, Francisco J. Beaumont, Javier Díez, Rick van Leeuwen, Robin Verjans, Leon J. De Windt, Blanche Schroen, Stephane Heymans, Chantal Munts, Cardiologie, RS: CARIM - R2.02 - Cardiomyopathy, RS: CARIM - R2 - Cardiac function and failure, Promovendi CD, Interne Geneeskunde, RS: CARIM - R3.01 - Vascular complications of diabetes and the metabolic syndrome, RS: CARIM - R2.07 - Gene regulation, Fysiologie, RS: CARIM - R2.08 - Electro mechanics, and MUMC+: MA Med Staf Spec Cardiologie (9)
- Subjects
0301 basic medicine ,Male ,TISSUE GROWTH-FACTOR ,heart failure ,LIVER FIBROSIS ,Cardiovascular Medicine ,Proto-Oncogene Mas ,Mice ,Fibrosis ,Transforming Growth Factor beta ,remodeling ,exercise ,General Commentary ,cardiac hypertrophy ,Dilated cardiomyopathy ,CARDIAC FIBROBLASTS ,micoRNAs ,microRNAs ,ANGIOTENSIN-II ,Signal Transduction ,cardiomyopathies ,TRANSFORMING GROWTH-FACTOR-BETA-1 ,medicine.medical_specialty ,BILIARY ATRESIA ,Diastole ,HEPATIC STELLATE CELLS ,SMOOTH MUSCLE ACTIN ,03 medical and health sciences ,Internal medicine ,fibroblasts ,Internal Medicine ,medicine ,Animals ,Humans ,AORTIC-STENOSIS ,INHIBITS AUTOPHAGY ,Pressure overload ,business.industry ,Myocardium ,Aortic Valve Stenosis ,medicine.disease ,Angiotensin II ,Rats ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Heart failure ,Hepatic stellate cell ,Myocardial fibrosis ,business - Abstract
Pressure overload causes cardiac fibroblast activation and transdifferentiation, leading to increased interstitial fibrosis formation and subsequently myocardial stiffness, diastolic and systolic dysfunction, and eventually heart failure. A better understanding of the molecular mechanisms underlying pressure overload-induced cardiac remodeling and fibrosis will have implications for heart failure treatment strategies. The microRNA (miRNA)-221/222 family, consisting of miR-221-3p and miR-222-3p, is differentially regulated in mouse and human cardiac pathology and inversely associated with kidney and liver fibrosis. We investigated the role of this miRNA family during pressure overload-induced cardiac remodeling. In myocardial biopsies of patients with severe fibrosis and dilated cardiomyopathy or aortic stenosis, we found significantly lower miRNA-221/222 levels as compared to matched patients with nonsevere fibrosis. In addition, miRNA-221/222 levels in aortic stenosis patients correlated negatively with the extent of myocardial fibrosis and with left ventricular stiffness. Inhibition of both miRNAs during AngII (angiotensin II)-mediated pressure overload in mice led to increased fibrosis and aggravated left ventricular dilation and dysfunction. In rat cardiac fibroblasts, inhibition of miRNA-221/222 derepressed TGF-beta (transforming growth factor-beta)-mediated profibrotic SMAD2 (mothers against decapentaplegic homolog 2) signaling and downstream gene expression, whereas overexpression of both miRNAs blunted TGF-beta-induced profibrotic signaling. We found that the miRNA-221/222 family may target several genes involved in TGF-beta signaling, including JNK1 (c-Jun N-terminal kinase 1), TGF-beta receptor 1 and TGF-beta receptor 2, and ETS-1 (ETS proto-oncogene 1). Our findings show that heart failure-associated downregulation of the miRNA-221/222 family enables profibrotic signaling in the pressure-overloaded heart.
- Published
- 2018
- Full Text
- View/download PDF
36. The innate immune system in chronic cardiomyopathy: a European Society of Cardiology (ESC) scientific statement from the Working Group on Myocardial Function of the ESC
- Author
-
Manuel Mayr, Mauro Giacca, Jolanda van der Velden, Denise Hilfiker-Kleiner, Inês Falcão-Pires, Thomas Thum, Adelino F. Leite-Moreira, Stephane Heymans, Emilio Hirsch, Dana Dawson, Leon J. De Windt, Johann Bauersachs, Michele Ciccarelli, Stefan Frantz, Gilda Varricchi, Carlo G. Tocchetti, Nazha Hamdani, Jean-Luc Balligand, and Dirk L. Brutsaert
- Subjects
0301 basic medicine ,Viral cardiomyopathy ,Innate immune system ,Peripartum cardiomyopathy ,business.industry ,T cell ,Cardiomyopathy ,biochemical phenomena, metabolism, and nutrition ,030204 cardiovascular system & hematology ,medicine.disease ,Bioinformatics ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Heart failure ,Diabetic cardiomyopathy ,medicine ,bacteria ,Cardiology and Cardiovascular Medicine ,business - Abstract
Activation of the immune system in heart failure (HF) has been recognized for over 20 years. Initially, experimental studies demonstrated a maladaptive role of the immune system. However, several phase III trials failed to show beneficial effects in HF with therapies directed against an immune activation. Preclinical studies today describe positive and negative effects of immune activation in HF. These different effects depend on timing and aetiology of HF. Therefore, herein we give a detailed review on immune mechanisms and their importance for the development of HF with a special focus on commonalities and differences between different forms of cardiomyopathies. The role of the immune system in ischaemic, hypertensive, diabetic, toxic, viral, genetic, peripartum, and autoimmune cardiomyopathy is discussed in depth. Overall, initial damage to the heart leads to disease specific activation of the immune system whereas in the chronic phase of HF overlapping mechanisms occur in different aetiologies.
- Published
- 2018
- Full Text
- View/download PDF
37. 3D microphysiological placenta in-vitro model for high-throughput barrier and transport studies
- Author
-
Chee Ng, Henriëtte L. Lanz, Leon J. De Windt, Gwenaëlle Rabussier, Kristin M. Bircsak, and Camilla Soragni
- Subjects
medicine.anatomical_structure ,Reproductive Medicine ,Chemistry ,Placenta ,medicine ,Obstetrics and Gynecology ,Throughput (business) ,Transport studies ,Developmental Biology ,In vitro model ,Cell biology - Published
- 2021
- Full Text
- View/download PDF
38. High throughput assay to quantify oxidative stress in organ-on-a-chip placenta models in a multi-chip platform
- Author
-
Sebastian J. Trietsch, Leon J. De Windt, Gwenaëlle Rabussier, Henriëtte L. Lanz, Camilla Soragni, and Chee Ping Ng
- Subjects
medicine.anatomical_structure ,Reproductive Medicine ,Chemistry ,Placenta ,medicine ,High Throughput Assay ,Obstetrics and Gynecology ,Chip ,medicine.disease_cause ,Organ-on-a-chip ,Oxidative stress ,Developmental Biology ,Cell biology - Published
- 2021
- Full Text
- View/download PDF
39. An integrative translational approach to study heart failure with preserved ejection fraction: a position paper from the Working Group on Myocardial Function of the European Society of Cardiology
- Author
-
Dana Dawson, Jean-Luc Balligand, Denise Hilfiker-Kleiner, Emilio Hirsch, Manuel Mayr, Christoph Maack, Burkert Pieske, Leon J. De Windt, Johann Bauersachs, Stefan Franz, Dirk L. Brutsaert, Thomas Thum, Inês Falcão-Pires, André P. Lourenço, Carlo G. Tocchetti, Stephane Heymans, Rudolf A. de Boer, Mauro Giacca, Adelino F. Leite-Moreira, and Ricardo Fontes-Carvalho
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Systems biology ,030204 cardiovascular system & hematology ,medicine.disease ,Myocardial function ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Animal model ,Internal medicine ,Heart failure ,medicine ,Cardiology ,Position paper ,In patient ,Translational science ,Cardiology and Cardiovascular Medicine ,Intensive care medicine ,Heart failure with preserved ejection fraction ,business - Abstract
As heart failure with preserved ejection fraction (HFpEF) rises to epidemic proportions, major steps in patient management and therapeutic development are badly needed. With the current position paper we seek to update our view on HFpEF as a highly complex systemic syndrome, from risk factors and mechanisms to long-term clinical manifestations. We will revise recent advances in animal model development, experimental set-ups and basic and translational science approaches to HFpEF research, highlighting their drawbacks and advantages. Directions are provided for proper model selection as well as for integrative functional evaluation from the in vivo setting to in vitro cell function testing. Additionally, we address new research challenges that require integration of higher-order inter-organ and inter-cell communication to achieve a full systems biology perspective of HFpEF.
- Published
- 2017
- Full Text
- View/download PDF
40. An unbiased silencing screen in muscle cells identifies miR-320a, miR-150, miR-196b, and miR-34c as regulators of skeletal muscle mitochondrial metabolism
- Author
-
Patrick Schrauwen, Joris Hoeks, Dennis Dahlmans, Johanna A. Jörgensen, Pénélope Andreux, Johan Auwerx, Leon J. De Windt, Alexandre Houzelle, Xu Wang, RS: NUTRIM - R1 - Metabolic Syndrome, Promovendi NTM, RS: NUTRIM - R1 - Obesity, diabetes and cardiovascular health, Cardiologie, RS: CARIM - R2.07 - Gene regulation, and RS: NUTRIM - HB/BW section B
- Subjects
Male ,EXPRESSION ,0301 basic medicine ,lcsh:Internal medicine ,MICRORNAS ,Muscle Fibers, Skeletal ,BIOGENESIS ,EXERCISE ,030209 endocrinology & metabolism ,Biology ,Mitochondrion ,DISEASE ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Skeletal muscle metabolism ,miR-150 ,microRNA ,medicine ,Animals ,Humans ,Gene silencing ,Myocyte ,lcsh:RC31-1245 ,Muscle, Skeletal ,PERSPECTIVE ,Molecular Biology ,INSULIN-RESISTANCE ,Myogenesis ,Skeletal muscle ,Cell Differentiation ,Myoblasts, Smooth Muscle ,MicroRNA ,Type 2 diabetes ,Cell Biology ,DYSFUNCTION ,Mitochondria, Muscle ,Mitochondria ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Biochemistry ,MIRNAS ,Screening ,Oxidative capacity ,Original Article ,HEALTH ,C2C12 - Abstract
Objective Strategies improving skeletal muscle mitochondrial capacity are commonly paralleled by improvements in (metabolic) health. We and others previously identified microRNAs regulating mitochondrial oxidative capacity, but data in skeletal muscle are limited. Therefore, the present study aimed to identify novel microRNAs regulating skeletal muscle mitochondrial metabolism. Methods and results We conducted an unbiased, hypothesis-free microRNA silencing screen in C2C12 myoblasts, using >700 specific microRNA inhibitors, and investigated a broad panel of mitochondrial markers. After subsequent validation in differentiated C2C12 myotubes, and exclusion of microRNAs without a human homologue or with an adverse effect on mitochondrial metabolism, 19 candidate microRNAs remained. Human clinical relevance of these microRNAs was investigated by measuring their expression in human skeletal muscle of subject groups displaying large variation in skeletal muscle mitochondrial capacity. Conclusion The results show that that microRNA-320a, microRNA-196b-3p, microRNA-150-5p, and microRNA-34c-3p are tightly related to in vivo skeletal muscle mitochondrial function in humans and identify these microRNAs as targets for improving mitochondrial metabolism., Graphical abstract Image 1, Highlights • A microRNA screen identifies microRNAs modulating muscle mitochondrial metabolism. • Identified microRNAs correlate with in vivo muscle mitochondrial function in humans. • MicroRNAs are potential targets to improve skeletal muscle mitochondrial metabolism.
- Published
- 2017
- Full Text
- View/download PDF
41. Wnt/β-catenin pathway in arrhythmogenic cardiomyopathy
- Author
-
Alessandra Rampazzo, Giulia Poloni, Alessandra Lorenzon, Martina Calore, Leon J. De Windt, and Paola Braghetta
- Subjects
0301 basic medicine ,RIGHT-VENTRICULAR CARDIOMYOPATHY ,NUCLEAR PLAKOGLOBIN ,Pathology ,medicine.medical_specialty ,Beta-catenin ,Cardiomyopathy ,Review ,030204 cardiovascular system & hematology ,Pathogenesis ,03 medical and health sciences ,Wnt ,0302 clinical medicine ,PRESSURE-OVERLOAD ,medicine ,WNT/FRIZZLED PATHWAY ,MAMMARY ONCOGENE INT-1 ,WNT SIGNALING PATHWAY ,Heart development ,biology ,INDUCED CARDIAC-HYPERTROPHY ,THERAPEUTIC TARGET ,business.industry ,Wnt signaling pathway ,beta-catenin ,β-catenin ,arrhythmogenic cardiomyopathy ,medicine.disease ,Molecular medicine ,microRNAs ,molecular pathogenesis ,030104 developmental biology ,Oncology ,Catenin ,biology.protein ,Cancer research ,Signal transduction ,business ,PLURIPOTENT STEM-CELLS - Abstract
// Alessandra Lorenzon 1 , Martina Calore 2 , Giulia Poloni 1 , Leon J. De Windt 2 , Paola Braghetta 3 and Alessandra Rampazzo 1 1 University of Padua, Department of Biology, Padua, Italy 2 Maastricht University, Department of Cardiology, Maastricht, The Netherlands 3 University of Padua, Department of Molecular Medicine, Padua, Italy Correspondence to: Alessandra Rampazzo, email: alessandra.rampazzo@unipd.it Keywords: Wnt, β-catenin, microRNAs, arrhythmogenic cardiomyopathy, molecular pathogenesis Received: January 30, 2017 Accepted: April 14, 2017 Published: April 27, 2017 ABSTRACT Wnt/β-catenin signaling pathway plays essential roles in heart development as well as cardiac tissue homoeostasis in adults. Abnormal regulation of this signaling pathway is linked to a variety of cardiac disease conditions, including hypertrophy, fibrosis, arrhythmias, and infarction. Recent studies on genetically modified cellular and animal models document a crucial role of Wnt/β-catenin signaling in the molecular pathogenesis of arrhythmogenic cardiomyopathy (AC), an inherited disease of intercalated discs, typically characterized by ventricular arrhythmias and progressive substitution of the myocardium with fibrofatty tissue. In this review, we summarize the conflicting published data regarding the Wnt/β-catenin signaling contribution to AC pathogenesis and we report the identification of a new potential therapeutic molecule that prevents myocyte injury and cardiac dysfunction due to desmosome mutations in vitro and in vivo by interfering in this signaling pathway. Finally, we underline the potential function of microRNAs, epigenetic regulatory RNA factors reported to participate in several pathological responses in heart tissue and in the Wnt signaling network, as important modulators of Wnt/β-catenin signaling transduction in AC. Elucidation of the precise regulatory mechanism of Wnt/β-catenin signaling in AC molecular pathogenesis could provide fundamental insights for new mechanism-based therapeutic strategy to delay the onset or progression of this cardiac disease.
- Published
- 2017
42. If you like it, put a ring on it!
- Author
-
Leon J. De Windt, Hamid el Azzouzi, RS: CARIM - R2.07 - Gene regulation, and Cardiologie
- Subjects
medicine.medical_specialty ,OUTFLOW TRACT OBSTRUCTION ,Physiology ,030204 cardiovascular system & hematology ,Ring (chemistry) ,Constriction ,Muscle hypertrophy ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Text mining ,Physiology (medical) ,Internal medicine ,Medicine ,Animals ,030212 general & internal medicine ,business.industry ,Heart ,Aortic Valve Stenosis ,medicine.disease ,HYPERTROPHY ,MODEL ,Phenotype ,Research Design ,Heart failure ,Aortic valve stenosis ,Cardiology ,HEART-FAILURE ,Cardiology and Cardiovascular Medicine ,business - Published
- 2018
43. Abstract 896: Cardiomyocyte-derived Mir-200c-3p In Exosomes Affects Endothelial Angiogenic Capacity And Impairs Cardiac Function
- Author
-
Diana S. Nascimento, Marida Sansonetti, Leon J. De Windt, Hamid el Azzouzi, Rio P. Juni, Vasco Sampaio-Pinto, Julie Halkein, Lara Ottaviani, Servé Olieslagers, and Paula A. da Costa Martins
- Subjects
Cardiac function curve ,Physiology ,Chemistry ,Mir 200c ,Cardiology and Cardiovascular Medicine ,Microvesicles ,Cell biology - Abstract
While cardiomyocytes (CMs) have been the main subject of extensive research, the role of other cardiac cell types, such as fibroblasts and endothelial cells (ECs), received considerable less attention in the pathogenesis of heart failure (HF). MiRNAs have recently emerged as mediators of paracrine signaling by being selectively incorporated in exosomes and exchanged between different cell types. The aim of our study is to investigate a potential paracrine miRNA crosstalk between CMs and cardiac ECs and assess the consequences of such miRNA transfer for cardiac vascular remodeling under pathological conditions. We isolated and characterized exosomes from CMs at baseline or after pathological stimulation with phenylephrine and isoproterenol. Although baseline and stressed CMs secrete miRNA-enriched exosomes at similar rates, comparative analysis of extracellular vesicles from both conditions revealed differential miRNA levels, with miR-200c-3p being highly enriched under stress conditions. Direct transfection of ECs with miR-200c-3p precursor molecules or indirect overexpression through transwell co-culture with stimulated CMs leads to diminished angiogenesis reflected by reduced capacity of ECs to proliferate, migrate, and form tubes. This effect was abrogated when we treated CMs with GW4869, an inhibitor of exosomal biogenesis and release. Next, we tested in vivo two doses of specific miR-200c-3p antagomir, Cy3 labelled, to assess specific target of ECs. FACS analysis on cardiac cells derived from the injected mice, confirmed that a low antagomir dose targets ECs whereas, the high dose of antagomir targets all different cardiac cell types. Moreover, when we treated mice subjected transverse aortic constriction (TAC)-induced cardiac pressure overload with miR-200c-3p antagomir, the animals developed a milder hypertrophic phenotype, smaller fibrotic areas, higher amount of capillaries and preserved cardiac ejection fraction, when compared to untreated pressure overloaded mice. Altogether, our results showing exosomal transfer of miR-200c-3p from CMs to ECs indicate the importance of cardiac intercellular communication in the pathophysiology of HF and identify a potential new therapeutic target for intervention strategies.
- Published
- 2019
- Full Text
- View/download PDF
44. Comparison of different chemically modified inhibitors of miR-199b in vivo
- Author
-
Rio P. Juni, Paula A. da Costa Martins, Lara Ottaviani, Burcu Duygu, Jan B. M. Wit, Leon J. De Windt, Nicole Bitsch, ACS - Heart failure & arrhythmias, Physiology, ACS - Microcirculation, RS: CARIM - R2 - Cardiac function and failure, Promovendi CD, Cardiologie, RS: FSE DMG, RS: CARIM - R2.07 - Gene regulation, Ondersteunend personeel CD, RS: Carim - H05 Gene regulation, and RS: CARIM School for Cardiovascular Diseases
- Subjects
0301 basic medicine ,EXPRESSION ,MICRORNAS ,IMPACT ,BIOGENESIS ,Mice, Inbred Strains ,Heart failure ,Pharmacology ,Biochemistry ,Chemical modifications ,OLIGONUCLEOTIDES ,DISEASE ,03 medical and health sciences ,chemistry.chemical_compound ,DELIVERY ,0302 clinical medicine ,Downregulation and upregulation ,In vivo ,TARGETS ,BIODISTRIBUTION ,microRNA ,medicine ,Animals ,Antagomir ,Antimirs ,Antisense oligonucleotides ,CELL-DIFFERENTIATION ,Dose-Response Relationship, Drug ,Cholesterol ,Oligonucleotide ,Antagomirs ,Heart ,medicine.disease ,030104 developmental biology ,Gene Expression Regulation ,chemistry ,030220 oncology & carcinogenesis ,Systemic administration ,Female - Abstract
MicroRNAs (miRNAs) have recently received great attention for their regulatory roles in diverse cellular processes and for their contribution to several human pathologies. Modulation of miRNAs in vivo provides beneficial therapeutic strategies for the treatment of many diseases, as evidenced by various preclinical studies. However, specific issues regarding the in vivo use of miRNA inhibitors (antimiRs) such as organ-specific delivery, optimal dosing and formulation of the best chemistry to obtain efficient miRNA inhibition remain to be addressed. Here, we aimed at comparing the in vivo efficacy of different chemistry-based antimiR oligonucleotides to inhibit cardiac expression of miR-199b, a highly promising therapeutic target for the treatment of pressure overload-induced cardiac dysfunction. For this purpose, four different designs of oligonucleotides to inhibit miR-199b were initially developed. Systemic administration to wildtype mice on three consecutive days was followed by organ harvesting, seven days after the first injection, in order to quantify the dose-dependent changes in miR-199b expression levels. When comparing the efficiency of each inhibitor at the highest applied dose we observed that the antagomir was the only inhibitor inducing complete inhibition of miR-199b in the heart. LNA reduced expression in the heart by 50 percent while the Zen-AMO and F/MOE chemistries failed to repress miR-199b expression in the heart at any given dose, in vivo. Further optimization was achieved by subjecting the antagomir and LNA nucleotides to additional chemical modifications. Interestingly, antagomir modification by replacing the cholesterol moiety from the 3′ to the 5′ end of the molecule significantly improved the inhibitory capacity, as reflected by a 75 percent downregulation of miR-199b expression already at a concentration of 5 mg/kg/day. Similar results could be obtained with a LNA-RNA molecule but upon administration of 80 mg/kg/day. These findings show that, from all the chemistries tested by us, an antagomir carrying the cholesterol group at the 5′ end was the most efficient inhibitor of miR-199b in the heart, in vivo. Moreover, our data also emphasize the importance of chemistry optimization and best dose range finding to achieve the greatest efficacy in miRNA inhibition in vivo.
- Published
- 2019
- Full Text
- View/download PDF
45. Therapeutic Delivery of miR-148a Suppresses Ventricular Dilation in Heart Failure
- Author
-
Mauro Giacca, Hamid el Azzouzi, Lorena Zentilin, Martinus I. F. J. Oerlemans, Natalia López-Andrés, Yigal M. Pinto, Paula A. da Costa Martins, Serena Zacchigna, Manon M. H. Huibers, Marida Sansonetti, Martina Calore, Andrea Raso, Roel A. de Weger, Leon J. De Windt, Amaya Fernández-Celis, Leonne E Philippen, Rio P. Juni, Nicole Bitsch, Federica De Majo, Servé Olieslagers, Ellen Dirkx, Vasco Sampaio-Pinto, Yolan J. Reckman, Raso, Andrea, Dirkx, Ellen, Philippen, Leonne E., Fernandez-Celis, Amaya, De Majo, Federica, Sampaio-Pinto, Vasco, Sansonetti, Marida, Juni, Rio, el Azzouzi, Hamid, Calore, Martina, Bitsch, Nicole, Olieslagers, Servé, Oerlemans, Martinus I. F. J., Huibers, Manon M., de Weger, Roel A., Reckman, Yolan J., Pinto, Yigal M., Zentilin, Lorena, Zacchigna, Serena, Giacca, Mauro, da Costa Martins, Paula A., López-Andrés, Natalia, De Windt, Leon J., Cardiologie, RS: CARIM - R2 - Cardiac function and failure, RS: CARIM - R2.07 - Gene regulation, Promovendi CD, RS: FSE DMG, Ondersteunend personeel CD, RS: Carim - Heart, RS: Carim - H05 Gene regulation, RS: CARIM School for Cardiovascular Diseases, Cardiology, Graduate School, ACS - Heart failure & arrhythmias, Physiology, and ACS - Microcirculation
- Subjects
adeno-associated vector ,cardiac ,heart failure ,hypertrophy ,microRNA ,miR-148a ,signaling ,Molecular Medicine ,Molecular Biology ,Genetics ,Pharmacology ,Drug Discovery3003 Pharmaceutical Science ,Volume overload ,Muscle hypertrophy ,Mice ,0302 clinical medicine ,Drug Discovery ,Medicine ,CARDIAC-HYPERTROPHY ,IN-VIVO ,0303 health sciences ,Ejection fraction ,Ventricular Remodeling ,GP130 ,Dilated cardiomyopathy ,3. Good health ,BRAIN NATRIURETIC PEPTIDE ,030220 oncology & carcinogenesis ,SURVIVAL ,Cardiology ,Original Article ,Cardiomyopathies ,Signal Transduction ,STAT3 Transcription Factor ,EXPRESSION ,INTERLEUKIN-6 FAMILY ,medicine.medical_specialty ,Concentric hypertrophy ,Cardiomegaly ,MYOCYTES ,03 medical and health sciences ,Genetic ,Internal medicine ,Journal Article ,Animals ,Humans ,PLASMA CARDIOTROPHIN-1 ,Ventricular remodeling ,Cell Proliferation ,030304 developmental biology ,Pressure overload ,business.industry ,Myocardium ,medicine.disease ,Dilatation ,GENE ,MicroRNAs ,Heart failure ,Commentary ,Heart Transplantation ,business - Abstract
Heart failure is preceded by ventricular remodeling, changes in left ventricular mass, and myocardial volume after alterations in loading conditions. Concentric hypertrophy arises after pressure overload, involves wall thickening, and forms a substrate for diastolic dysfunction. Eccentric hypertrophy develops in volume overload conditions and leads wall thinning, chamber dilation, and reduced ejection fraction. The molecular events underlying these distinct forms of cardiac remodeling are poorly understood. Here, we demonstrate that miR-148a expression changes dynamically in distinct subtypes of heart failure: while it is elevated in concentric hypertrophy, it decreased in dilated cardiomyopathy. In line, antagomir-mediated silencing of miR-148a caused wall thinning, chamber dilation, increased left ventricle volume, and reduced ejection fraction. Additionally, adeno-associated viral delivery of miR-148a protected the mouse heart from pressure-overload-induced systolic dysfunction by preventing the transition of concentric hypertrophic remodeling toward dilation. Mechanistically, miR-148a targets the cytokine co-receptor glycoprotein 130 (gp130) and connects cardiomyocyte responsiveness to extracellular cytokines by modulating the Stat3 signaling. These findings show the ability of miR-148a to prevent the transition of pressure-overload induced concentric hypertrophic remodeling toward eccentric hypertrophy and dilated cardiomyopathy and provide evidence for the existence of separate molecular programs inducing distinct forms of myocardial remodeling., Raso et al. show that miR-148a is elevated in concentric hypertrophy and decreased in dilated cardiomyopathy. Adeno-associated viral delivery of miR-148a protects the mouse heart from cardiac dilation. Mechanistically, miR-148a regulates the sensitivity of the heart to extracellular cytokines.
- Published
- 2019
- Full Text
- View/download PDF
46. Why publish in the American Journal of Physiology-Heart and Circulatory Physiology?
- Author
-
Leon J. De Windt, David A. Kass, Debra I. Diz, Christine Des Rosiers, Junichi Sadoshima, Crystal M. Ripplinger, Ronglih Liao, Kaushik P. Patel, Nisha Charkoudian, Donal S. O'Leary, Gary D. Lopaschuk, Mario Delmar, Fabio A. Recchia, Michael S. Wolin, Igor R. Efimov, Steven P. Jones, Robert L. Hester, Zoltan Ungvari, Nancy L. Kanagy, Ivor J. Benjamin, Merry L. Lindsey, Irving H. Zucker, Ajay M. Shah, Mordecai P. Blaustein, Yulin Liao, David D. Gutterman, Masafumi Kitakaze, RS: CARIM - R2.07 - Gene regulation, and Cardiologie
- Subjects
0301 basic medicine ,business.industry ,Physiology ,education ,MEDLINE ,030204 cardiovascular system & hematology ,Cardiology and Cardiovascular Medicine ,Physiology (medical) ,humanities ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Circulatory system ,Medicine ,Cardiovascular Physiological Phenomena ,Early career ,business - Abstract
for senior cardiovascular scientists, it is well known and accepted that the American Journal of Physiology ( AJP ) -Heart and Circulatory Physiology publishes important and long-lasting cardiovascular science ([2][1]). Early career investigators and trainees may be less familiar with the history of
- Published
- 2017
- Full Text
- View/download PDF
47. RNA therapeutics for heart disease
- Author
-
Federica De Majo, Leon J. De Windt, Promovendi CD, RS: CARIM - R2.07 - Gene regulation, and Cardiologie
- Subjects
0301 basic medicine ,RNA, Untranslated ,Heart disease ,Heart Diseases ,Druggability ,INHIBITION ,Computational biology ,030204 cardiovascular system & hematology ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Oligonucleotide ,PRESSURE-OVERLOAD ,medicine ,Animals ,Humans ,FAILURE ,Functional studies ,RNA, Small Interfering ,Pharmacology ,Gene Editing ,IMPROVES CARDIAC-FUNCTION ,RNA ,RNA therapeutics ,NONHUMAN-PRIMATES ,Translation (biology) ,Genetic Therapy ,CARDIOMYOCYTE APOPTOSIS ,Chemical basis ,medicine.disease ,3. Good health ,HYPERTROPHY ,MicroRNAs ,MICE ,030104 developmental biology ,MYOCARDIAL-INFARCTION ,NONCODING RNAS ,Drug Design ,RNA-based drugs ,Human genome ,RNA, Long Noncoding - Abstract
The majority of the human genome encodes non-coding RNAs (ncRNAs), species of RNA without protein-coding potential but with powerful regulatory functions in disease onset and progression. Functional studies demonstrate that both coding and ncRNAs underlie various mechanisms in heart disease and that molecules targeting RNA species show promising efficacy in preclinical development. Accompanying the exciting developments in basic RNA biology, an equally provocative field has flourished for the design of RNA-based strategies to generate innovative types of therapeutics against these new "druggable" targets, going beyond our current repertoire of small chemistry or biologics. Here, we review the (bio)chemical basis of RNA-based drug design, provide examples that show promise as translatable drug products in preclinical studies, give an insight in the current barriers that hamper straight-forward clinical translation and discuss future directions that may overcome these hurdles to expand the current pharmacotherapy for myocardial disorders.
- Published
- 2018
48. The innate immune system in chronic cardiomyopathy:a European Society of Cardiology (ESC) scientific statement from the Working Group on Myocardial Function of the ESC
- Author
-
Stefan, Frantz, Ines, Falcao-Pires, Jean-Luc, Balligand, Johann, Bauersachs, Dirk, Brutsaert, Michele, Ciccarelli, Dana, Dawson, Leon J, de Windt, Mauro, Giacca, Nazha, Hamdani, Denise, Hilfiker-Kleiner, Emilio, Hirsch, Adelino, Leite-Moreira, Manuel, Mayr, Thomas, Thum, Carlo G, Tocchetti, Jolanda, van der Velden, Gilda, Varricchi, Stephane, Heymans, Frantz, Stefan, Falcao-Pires, Ine, Balligand, Jean-Luc, Bauersachs, Johann, Brutsaert, Dirk, Ciccarelli, Michele, Dawson, Dana, de Windt, Leon J., Giacca, Mauro, Hamdani, Nazha, Hilfiker-Kleiner, Denise, Hirsch, Emilio, Leite-Moreira, Adelino, Mayr, Manuel, Thum, Thoma, Tocchetti, Carlo G., van der Velden, Jolanda, Varricchi, Gilda, and Heymans, Stephane
- Subjects
Autoimmune cardiomyopathy ,Diabetic cardiomyopathy ,Genetic cardiomyopathy ,Hypertensive cardiomyopathy ,Immune system ,Ischaemic cardiomyopathy ,Macrophage ,Peripartum cardiomyopathy ,T-cell ,Toxic cardiomyopathy ,Viral cardiomyopathy ,Cardiomyopathies ,Heart Failure ,Humans ,Myocardium ,Stroke Volume ,Disease Progression ,Immunity, Innate ,Registries ,Innate ,Immunity ,biochemical phenomena, metabolism, and nutrition ,Cardiology and Cardiovascular Medicine ,bacteria ,Human medicine - Abstract
Activation of the immune system in heart failure (HF) has been recognized for over 20 years. Initially, experimental studies demonstrated a maladaptive role of the immune system. However, several phase III trials failed to show beneficial effects in HF with therapies directed against an immune activation. Preclinical studies today describe positive and negative effects of immune activation in HF. These different effects depend on timing and aetiology of HF. Therefore, herein we give a detailed review on immune mechanisms and their importance for the development of HF with a special focus on commonalities and differences between different forms of cardiomyopathies. The role of the immune system in ischaemic, hypertensive, diabetic, toxic, viral, genetic, peripartum, and autoimmune cardiomyopathy is discussed in depth. Overall, initial damage to the heart leads to disease specific activation of the immune system whereas in the chronic phase of HF overlapping mechanisms occur in different aetiologies.
- Published
- 2018
- Full Text
- View/download PDF
49. Antisense MicroRNA Therapeutics in Cardiovascular Disease: Quo Vadis?
- Author
-
Jan B. M. Wit, Leonne E Philippen, Ellen Dirkx, Koos Burggraaf, Leon J. De Windt, Paula A. da Costa Martins, RS: CARIM - R2 - Cardiac function and failure, Promovendi CD, and Cardiologie
- Subjects
Review ,Disease ,Pharmacology ,Bioinformatics ,Sudden death ,Coronary artery disease ,microRNA ,Drug Discovery ,Genetics ,Animals ,Humans ,Medicine ,In patient ,Pathological ,Molecular Biology ,Heart Failure ,business.industry ,Myocardium ,medicine.disease ,3. Good health ,Disease Models, Animal ,MicroRNAs ,Antisense Elements (Genetics) ,Tissue remodeling ,Gene Expression Regulation ,Heart failure ,Molecular Medicine ,business - Abstract
Heart failure (HF) is the end result of a diverse set of causes such as genetic cardiomyopathies, coronary artery disease, and hypertension and represents the primary cause of hospitalization in Europe. This serious clinical disorder is mostly associated with pathological remodeling of the myocardium, pump failure, and sudden death. While the survival of HF patients can be prolonged with conventional pharmacological therapies, the prognosis remains poor. New therapeutic modalities are thus needed that will target the underlying causes and not only the symptoms of the disease. Under chronic cardiac stress, small noncoding RNAs, in particular microRNAs, act as critical regulators of cardiac tissue remodeling and represent a new class of therapeutic targets in patients suffering from HF. Here, we focus on the potential use of microRNA inhibitors as a new treatment paradigm for HF.
- Published
- 2015
- Full Text
- View/download PDF
50. Non-coding RNA in control of gene regulatory programs in cardiac development and disease
- Author
-
Leonne E Philippen, Paula A da Costa-Martins, Leon J. De Windt, Ellen Dirkx, Promovendi CD, Cardiologie, and RS: CARIM - R2 - Cardiac function and failure
- Subjects
Genetics ,Regulation of gene expression ,Gene regulatory network ,Gene Expression Regulation, Developmental ,Heart ,Computational biology ,Biology ,Non-coding RNA ,Embryonic stem cell ,Gene Expression Regulation ,Cardiovascular Diseases ,microRNA ,Gene expression ,RNA ,Animals ,Humans ,Developmental ,Long Noncoding ,Gene Regulatory Networks ,RNA, Long Noncoding ,Epigenetics ,Cardiology and Cardiovascular Medicine ,Molecular Biology ,Gene - Abstract
Organogenesis of the vertebrate heart is a highly specialized process involving progressive specification and differentiation of distinct embryonic cardiac progenitor cell populations driven by specialized gene programming events. Likewise, the onset of pathologies in the adult heart, including cardiac hypertrophy, involves the reactivation of embryonic gene programs. In both cases, these intricate genomic events are temporally and spatially regulated by complex signaling networks and gene regulatory networks. Apart from well-established transcriptional mechanisms, increasing evidence indicates that gene programming in both the developing and the diseased myocardium are under epigenetic control by non-coding RNAs (ncRNAs). MicroRNAs regulate gene expression at the post-transcriptional level, and numerous studies have now established critical roles for this species of tiny RNAs in a broad range of aspects from cardiogenesis towards adult heart failure. Recent reports now also implicate the larger family of long non-coding RNAs (lncRNAs) in these processes as well. Here we discuss the involvement of these two ncRNA classes in proper cardiac development and hypertrophic disease processes of the adult myocardium. This article is part of a Special Issue entitled: Non-coding RNAs.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.